1
|
Rabie MA, Ghoneim AT, Fahmy MI, El-Yamany MF, Sayed RH. Activation of alpha-7 nicotinic acetylcholine receptor by tropisetron mitigates 3-nitropropionic acid-induced Huntington's disease in rats: Role of PI3K/Akt and JAK2/NF-κB signaling pathways. Chem Biol Interact 2024; 393:110957. [PMID: 38513929 DOI: 10.1016/j.cbi.2024.110957] [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: 01/30/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Huntington's disease (HD) is an inheritable autosomal-dominant disorder that targets mainly the striatum. 3-Nitropropionic acid (3-NP) induces obvious deleterious behavioral, neurochemical, and histological effects similar to the symptoms of HD. Our study aimed to examine the neuroprotective activity of tropisetron, an alpha-7 neuronal nicotinic acetylcholine receptor (α-7nAChR) agonist, against neurotoxic events associated with 3-NP-induced HD in rats. Forty-eight rats were randomly allocated into four groups. Group I received normal saline, while Groups II, III and IV received 3-NP for 2 weeks. In addition, Group III and IV were treated with tropisetron 1 h after 3-NP administration. Meanwhile, Group IV received methyllycaconitine (MLA), an α-7nAChR antagonist, 30 min before tropisetron administration. Treatment with tropisetron improved motor deficits as confirmed by the behavioral tests and restored normal histopathological features of the striatum. Moreover, tropisetron showed an anti-oxidant activity via increasing the activities of SDH and HO-1 as well as Nrf2 expression along with reducing MDA level. Tropisetron also markedly upregulated the protein expression of p-PI3K and p-Akt which in turn hampered JAK2/NF-κB inflammatory cascade. In addition, tropisetron showed an anti-apoptotic activity through boosting the expression of Bcl-2 and reducing Bax expression and caspase-3 level. Interestingly, all the aforementioned effects of tropisetron were blocked by pre-administration of MLA, which confirms that such neuroprotective effects are mediated via activating of α-7nAChR. In conclusion, tropisetron showed a neuroprotective activity against 3-NP-induced HD via activating PI3K/Akt signaling and suppressing JAK2/NF-κB inflammatory axis. Thus, repositioning of tropisetron could represent a promising therapeutic strategy in management of HD.
Collapse
Affiliation(s)
- Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Ahmed T Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical sciences and drug manufacturing, Misr University for Science and Technology (MUST), 12585, Giza, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt; School of Pharmacy, Newgiza University, Giza, Egypt
| |
Collapse
|
2
|
Guzman-Vallejos MS, Ramirez-Cando LJ, Aguayo L, Ballaz SJ. Molecular Docking Analysis at the Human α7-nAChR and Proliferative and Evoked-Calcium Changes in SH-SY5Y Cells by Imidacloprid and Acetamiprid Insecticides. Neurotox Res 2024; 42:16. [PMID: 38376791 DOI: 10.1007/s12640-024-00697-0] [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: 11/06/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Acetamiprid (ACE) and Imidacloprid (IMI) are widely-used neonicotinoid insecticides (NNIs) with functional activity at human acetylcholine nicotinic receptors and, therefore, with putative toxic effects. The objective of this study was the evaluation of the interactions between NNIs and α7-nAChR, as this receptor keeps intracellular Ca2+ ([Ca2+]i) to an optimum for an adequate neuronal functioning. Possible interactions between NNIs and the cryo-EM structure of the human α-7 nAChR were identified by molecular docking. Additionally, NNI effects were analyzed in neuroblastoma SH-SY5Y cells, as they naturally express α-7 nAChRs. Functional studies included proliferative/cytotoxic effects (MTT test) in undifferentiated SH-SY-5Y cells and indirect measurements of [Ca2+]i transients in retinoic acid-differentiated SH-SY-5Y cells loaded with Fluo-4 AM. Docking analysis showed that the binding of IMI and ACE occurred at the same aromatic cage that the specific α-7 nAChR agonist EVP-6124. IMI showed a better docking strength than ACE. According to the MTT assays, low doses (10-50 µM) of IMI better than ACE stimulated neuroblastoma cell proliferation. At higher doses (250-500 µM), IMI also prevailed over ACE and dose-dependently triggered more abrupt fluorescence changes due to [Ca2+]i mobilization in differentiated SH-SY5Y neurons. Indeed, only IMI blunted nicotine-evoked intracellular fluorescence stimulation (i.e., nicotine cross-desensitization). Summarizing, IMI demonstrated a superior docking strength and more robust cellular responses compared to ACE, which were likely associated with a stronger activity at α-7nAChRs. Through the interaction with α-7nAChRs, IMI would demonstrate its high neurotoxic potential for humans. More research is needed for investigating the proliferative effects of IMI in neuroblastoma cells.
Collapse
Affiliation(s)
| | - Lenin J Ramirez-Cando
- School of Biological Sciences & Engineering, Universidad Yachay Tech, Urcuquí, Ecuador
| | - Luis Aguayo
- School of Biological Sciences, Universidad de Concepcion, Concepción, Chile
| | - Santiago J Ballaz
- School of Medicine, Universidad Espíritu Santo, Ave. Samborondón 5, Samborondón, 0901952, Ecuador.
| |
Collapse
|
3
|
Singh S, Agrawal N, Goyal A. Role of Alpha-7-Nicotinic Acetylcholine Receptor in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:384-394. [PMID: 37366362 DOI: 10.2174/1871527322666230627123426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder affecting millions worldwide. One of the leading hypotheses for the underlying cause of AD is a reduction in nicotinic receptor levels in the brain. Among the nicotinic receptors, the alpha-7-nicotinic acetylcholine receptor (α7nAChR) has received particular attention due to its involvement in cognitive function.α7nAChR is a ligand-gated ion channel that is primarily found in the hippocampus and prefrontal cortex, areas of the brain responsible for learning, memory, and attention. Studies have shown that α7nAChR dysfunction is a key contributor to the pathogenesis of AD. The receptor is involved in regulating amyloidbeta (Aβ) production, a hallmark of AD pathology. Many drugs have been investigated as α7nAChR agonists or allosteric modulators to improve cognitive deficits in AD. Clinical studies have shown promising results with α7nAChR agonists, including improved memory and cognitive function. Although several studies have shown the significance of the α7 nAChR in AD, little is known about its function in AD pathogenesis. As a result, in this review, we have outlined the basic information of the α7 nAChR's structure, functions, cellular responses to its activation, and its role in AD's pathogenesis.
Collapse
Affiliation(s)
- Sushma Singh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
- Pharmacy College, Azamgarh- 276128, UP, India
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| |
Collapse
|
4
|
Echeverria V, Mendoza C, Iarkov A. Nicotinic acetylcholine receptors and learning and memory deficits in Neuroinflammatory diseases. Front Neurosci 2023; 17:1179611. [PMID: 37255751 PMCID: PMC10225599 DOI: 10.3389/fnins.2023.1179611] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/07/2023] [Indexed: 06/01/2023] Open
Abstract
Animal survival depends on cognitive abilities such as learning and memory to adapt to environmental changes. Memory functions require an enhanced activity and connectivity of a particular arrangement of engram neurons, supported by the concerted action of neurons, glia, and vascular cells. The deterioration of the cholinergic system is a common occurrence in neurological conditions exacerbated by aging such as traumatic brain injury (TBI), posttraumatic stress disorder (PTSD), Alzheimer's disease (AD), and Parkinson's disease (PD). Cotinine is a cholinergic modulator with neuroprotective, antidepressant, anti-inflammatory, antioxidant, and memory-enhancing effects. Current evidence suggests Cotinine's beneficial effects on cognition results from the positive modulation of the α7-nicotinic acetylcholine receptors (nAChRs) and the inhibition of the toll-like receptors (TLRs). The α7nAChR affects brain functions by modulating the function of neurons, glia, endothelial, immune, and dendritic cells and regulates inhibitory and excitatory neurotransmission throughout the GABA interneurons. In addition, Cotinine acting on the α7 nAChRs and TLR reduces neuroinflammation by inhibiting the release of pro-inflammatory cytokines by the immune cells. Also, α7nAChRs stimulate signaling pathways supporting structural, biochemical, electrochemical, and cellular changes in the Central nervous system during the cognitive processes, including Neurogenesis. Here, the mechanisms of memory formation as well as potential mechanisms of action of Cotinine on memory preservation in aging and neurological diseases are discussed.
Collapse
Affiliation(s)
- Valentina Echeverria
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Research and Development Department, Bay Pines VAHCS, Bay Pines, FL, United States
| | - Cristhian Mendoza
- Facultad de Odontologia y Ciencias de la Rehabilitacion, Universidad San Sebastián, Concepción, Chile
| | - Alex Iarkov
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| |
Collapse
|
5
|
α7nAChR activation protects against oxidative stress, neuroinflammation and central insulin resistance in ICV-STZ induced sporadic Alzheimer's disease. Pharmacol Biochem Behav 2022; 217:173402. [DOI: 10.1016/j.pbb.2022.173402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 12/29/2022]
|
6
|
Chavda V, Singh K, Patel V, Mishra M, Mishra AK. Neuronal Glial Crosstalk: Specific and Shared Mechanisms in Alzheimer’s Disease. Brain Sci 2022; 12:brainsci12010075. [PMID: 35053818 PMCID: PMC8773743 DOI: 10.3390/brainsci12010075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The human brain maintains billions of neurons functional across the lifespan of the individual. The glial, supportive cells of the brain are indispensable to neuron elasticity. They undergo various states (active, reactive, macrophage, primed, resting) and carefully impose either quick repair or the cleaning of injured neurons to avoid damage extension. Identifying the failure of these interactions involving the relation of the input of glial cells to the inception and/or progression of chronic neurodegenerative diseases (ND) is crucial in identifying therapeutic options, given the well-built neuro-immune module of these diseases. In the present review, we scrutinize different interactions and important factors including direct cell–cell contact, intervention by the CD200 system, various receptors present on their surfaces, CXC3RI and TREM2, and chemokines and cytokines with special reference to Alzheimer’s disease (AD). The present review of the available literature will elucidate the contribution of microglia and astrocytes to the pathophysiology of AD, thus evidencing glial cells as obligatory transducers of pathology and superlative targets for interference.
Collapse
Affiliation(s)
- Vishal Chavda
- Division of Anesthesia, Dreamzz IVF Center and Women’s Care Hospital, Ahmedabad 382350, Gujarat, India;
| | - Kavita Singh
- Centre for Translational Research, Jiwaji University, Gwalior 474011, Madhya Pradesh, India;
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Meerambika Mishra
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (M.M.); (A.K.M.)
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Korea
- Correspondence: (M.M.); (A.K.M.)
| |
Collapse
|
7
|
Patro N, Kushwaha SS, Patro I. Microglia Aging. THE BIOLOGY OF GLIAL CELLS: RECENT ADVANCES 2022:565-592. [DOI: 10.1007/978-981-16-8313-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Tsai ST, Wei TH, Yang YW, Lu MK, San S, Tsai CH, Lin YW. Transient receptor potential V1 modulates neuroinflammation in Parkinson's disease dementia: Molecular implications for electroacupuncture and rivastigmine. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1336-1345. [PMID: 35096291 PMCID: PMC8769514 DOI: 10.22038/ijbms.2021.56156.12531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/17/2021] [Indexed: 12/05/2022]
Abstract
OBJECTIVES Parkinson's disease (PD) is a common progressive neurodegeneration disease. Its incidence increases with age and affects about 1% of people over 60. Incidentally, transient receptor potential V1 (TRPV1) and its relation with neuroinflammation in mouse brain has been widely reported. MATERIALS AND METHODS We used 6-hydroxydopamine (6-OHDA) to induce PDD in mice. We then used the Morris water maze and Bio-Plex to test learning and inflammatory mediators in mouse plasma. Western blotting and immunostaining were used to examine TRPV1 pathway in the hippocampus and medial prefrontal cortex (mPFC). RESULTS On acquisition days 3 (Control = 4.40 ± 0.8 sec, PDD = 9.82 ± 1.52 sec, EA = 5.04 ± 0.58 sec, Riva = 4.75 ± 0.87 sec; P=0.001) and 4, reversal learning days 1, 2, 3 (Control = 2.86 ± 0.46 sec, PDD = 9.80 ± 1.83 sec, EA = 4.6 ± 0.82 sec, Riva = 4.6 ± 1.03 sec; P=0.001) and 4, PDD mice showed significantly longer escape latency than the other three groups. Results showed that several cytokines were up-regulated in PDD mice and reversed by EA and rivastigmine. TRPV1 and downstream molecules were up-regulated in PDD mice and further reversed by EA and rivastigmine. Interestingly, α7 nicotinic receptors and parvalbumin levels in both the hippocampus and prefrontal cortex increased in EA-treated mice, but not in rivastigmine-treated mice. CONCLUSION Our results showed that TRPV1 played a role in the modulation of neuroinflammation of PDD, and could potentially be a new target for treatment.
Collapse
Affiliation(s)
- Sheng-Ta Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan, Everflourish Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Hsuan Wei
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Wan Yang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Kuei Lu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Everflourish Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shao San
- Department of Psychiatry, Taoyuan Psychiatric Center, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Everflourish Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan, College of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Wen Lin
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan, Chinese Medicine Research Center, China Medical University, Taichung, Taiwan,Corresponding author: Yi-Wen Lin. Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No. 91, Xueshi Road, North District, Taichung 404, Taiwan. Tel: 886-970055508;
| |
Collapse
|
9
|
Zhao X, Wilson K, Uteshev V, He JJ. Activation of α7 nicotinic acetylcholine receptor ameliorates HIV-associated neurology and neuropathology. Brain 2021; 144:3355-3370. [PMID: 34196664 PMCID: PMC8677536 DOI: 10.1093/brain/awab251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/28/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
HIV-associated neurocognitive disorders (HAND) in the era of combination antiretroviral therapy are primarily manifested as impaired behaviours, glial activation/neuroinflammation and compromised neuronal integrity, for which there are no effective treatments currently available. In the current study, we used doxycycline-inducible astrocyte-specific HIV Tat transgenic mice (iTat), a surrogate HAND model, and determined effects of PNU-125096, a positive allosteric modulator of α7 nicotinic acetylcholine receptor (α7 nAChR) on Tat-induced behavioural impairments and neuropathologies. We showed that PNU-125096 treatment significantly improved locomotor, learning and memory deficits of iTat mice while inhibited glial activation and increased PSD-95 expression in the cortex and hippocampus of iTat mice. Using α7 nAChR knockout mice, we showed that α7 nAChR knockout eliminated the protective effects of PNU-125096 on iTat mice. In addition, we showed that inhibition of p38 phosphorylation by SB239063, a p38 MAPK-specific inhibitor exacerbated Tat neurotoxicity in iTat mice. Last, we used primary mouse cortical individual cultures and neuron-astrocytes co-cultures and in vivo staining of iTat mouse brain tissues and showed that glial activation was directly involved in the interplay among Tat neurotoxicity, α7 nAChR activation and the p38 MAPK signalling pathway. Taken together, these findings demonstrated for the first time that α7 nAChR activation led to protection against HAND and suggested that α7 nAChR modulator PNU-125096 holds significant promise for development of therapeutics for HAND.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Kelly Wilson
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Victor Uteshev
- Department of Pharmacology and Neuroscience, Graduate School of Biomedical Sciences of University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Johnny J He
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| |
Collapse
|
10
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
11
|
Spatiotemporal Patterns of Menin Localization in Developing Murine Brain: Co-Expression with the Elements of Cholinergic Synaptic Machinery. Cells 2021; 10:cells10051215. [PMID: 34065662 PMCID: PMC8156519 DOI: 10.3390/cells10051215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/02/2021] [Accepted: 05/10/2021] [Indexed: 01/05/2023] Open
Abstract
Menin, a product of MEN1 (multiple endocrine neoplasia type 1) gene is an important regulator of tissue development and maintenance; its perturbation results in multiple tumors—primarily of the endocrine tissue. Despite its abundance in the developing central nervous system (CNS), our understanding of menin’s role remains limited. Recently, we discovered menin to play an important role in cholinergic synaptogenesis in the CNS, whereas others have shown its involvement in learning, memory, depression and apoptosis. For menin to play these important roles in the CNS, its expression patterns must be corroborated with other components of the synaptic machinery imbedded in the learning and memory centers; this, however, remains to be established. Here, we report on the spatio-temporal expression patterns of menin, which we found to exhibit dynamic distribution in the murine brain from early development, postnatal period to a fully-grown adult mouse brain. We demonstrate here that menin expression is initially widespread in the brain during early embryonic stages, albeit with lower intensity, as determined by immunohistochemistry and gene expression. With the progression of development, however, menin expression became highly localized to learning, memory and cognition centers in the CNS. In addition to menin expression patterns throughout development, we provide the first direct evidence for its co-expression with nicotinic acetylcholine, glutamate and GABA (gamma aminobutyric acid) receptors—concomitant with the expression of both postsynaptic (postsynaptic density protein PSD-95) and presynaptic (synaptotagamin) proteins. This study is thus the first to provide detailed analysis of spatio-temporal patterns of menin expression from initial CNS development to adulthood. When taken together with previously published studies, our data underscore menin’s importance in the cholinergic neuronal network assembly underlying learning, memory and cognition.
Collapse
|
12
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
13
|
Jiang Y, Ma H, Wang X, Wang Z, Yang Y, Li L, Feng T. Protective Effect of the α7 Nicotinic Receptor Agonist PNU-282987 on Dopaminergic Neurons Against 6-Hydroxydopamine, Regulating Anti-neuroinflammatory and the Immune Balance Pathways in Rat. Front Aging Neurosci 2021; 12:606927. [PMID: 33568987 PMCID: PMC7868536 DOI: 10.3389/fnagi.2020.606927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/18/2020] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation and inner immune dysfunction are increasingly accepted as important components of the etiopathogenesis of Parkinson’s disease (PD). According to emerging evidence, a7 nicotinic acetylcholine receptor (α7nAChR), a ligand-gated ion channel, plays an important role in inflammatory reactions and is also expressed on the surface of T cells. In particular, regulatory T cells (Tregs) are critical for the maintenance of immunological tolerance. In the present study, we investigated the roles of α7nAChR in inhibiting inflammation and maintaining the immune balance in rats with 6-hydroxydopamine (6-OHDA)-induced lesions and the possible mechanisms regulating the proportion of Tregs in vivo. Adult male Wistar rats (n = 90) were subjected to a unilateral injection of 6-OHDA into the left medial forebrain bundle, and PNU-282987, an α7nAChR agonist, was intraperitoneally injected 2 h prior to the induction of lesions by 6-OHDA and again at days 1, 7, and 13 postlesion. Behavioral tests and immunohistochemical staining to detect the expression of tyrosine hydroxylase (TH) in the bilateral substantial nigra (SN) were performed. Subsequently, CD4+ T lymphocytes and the expression of forkhead/winged helix transcription factor p3 (Foxp3, which is a marker of Treg cells) in the SN were also assessed using immunofluorescence staining. The expression of glial fibrillary acidic protein (GFAP) in the SN was determined by performing immunohistochemical staining. Additionally, the protein levels of α7nAChR, extracellular signal-regulated kinase (Erk) phosphorylated-Erk (p-Erk) and Foxp3 in the ventral midbrain were determined using Western blotting, and the relative expression of the TNF-α, IL-1β, and IL-10 mRNAs were detected using real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). We found that PNU-282987 significantly improved the motor deficits induced by 6-OHDA, reduced the loss of TH in the SN, suppressed the overactivation of GFAP+ cells and expression of related inflammatory cytokines, and increased the number of Foxp3+ cells. In addition, we also showed that PNU-282987 significantly increased the protein expression of the a7nAchR, p-Erk, and Foxp3 in 6-OHDA-lesioned rats (p < 0.05). These results indicated that α7nAChR activation could exert an anti-inflammatory effect and participate in the process of modulating the immune balance during 6-OHDA-induced injury, potentially through the α7nAChR/p-Erk/Foxp3 signaling pathway.
Collapse
Affiliation(s)
- Ying Jiang
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huizi Ma
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xuemei Wang
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhan Wang
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yaqin Yang
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Longling Li
- Department of Neurology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Tao Feng
- Center for Movement Disorders Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
14
|
Changing Functional Signatures of Microglia along the Axis of Brain Aging. Int J Mol Sci 2021; 22:ijms22031091. [PMID: 33499206 PMCID: PMC7865559 DOI: 10.3390/ijms22031091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia, the innate immune cells of the brain, are commonly perceived as resident macrophages of the central nervous system (CNS). This definition, however, requires further specification, as under healthy homeostatic conditions, neither morphological nor functional properties of microglia mirror those of classical macrophages. Indeed, microglia adapt exceptionally well to their microenvironment, becoming a legitimate member of the cellular brain architecture. The ramified or surveillant microglia in the young adult brain are characterized by specific morphology (small cell body and long, thin motile processes) and physiology (a unique pattern of Ca2+ signaling, responsiveness to various neurotransmitters and hormones, in addition to classic “immune” stimuli). Their numerous physiological functions far exceed and complement their immune capabilities. As the brain ages, the respective changes in the microglial microenvironment impact the functional properties of microglia, triggering further rounds of adaptation. In this review, we discuss the recent data showing how functional properties of microglia adapt to age-related changes in brain parenchyma in a sex-specific manner, with a specific focus on early changes occurring at middle age as well as some strategies counteracting the aging of microglia.
Collapse
|
15
|
Rogers SW, Myers EJ, Gahring LC. Age-Associated Tooth Loss and Oral Microbial Dysbiosis in a Mouse Genetic Model of Chronic Nicotine Exposure. Front Immunol 2020; 11:575200. [PMID: 33117372 PMCID: PMC7575759 DOI: 10.3389/fimmu.2020.575200] [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: 06/22/2020] [Accepted: 09/08/2020] [Indexed: 11/13/2022] Open
Abstract
Nicotine acts as a potent modulator of normal cellular responses through the nicotinic acetylcholine receptor subtype alpha7. In a mouse genetic model of alpha7 receptor dysfunction, alpha7E260A:G, 85 percent of 18 month-old mice exhibit an age-associated spontaneous loosening or complete loss of 3rd molars that was not present in the control mice. The adjacent soft tissues appeared largely unaffected. Further analysis including micro-CT revealed evidence of bone loss surrounding the 3rd molars with areas of cavitation and/or sponge-like (cancellous) bone remodeling in the mandible. The mandible microbiome was examined using 16S-rRNA sequencing. The results show the alpha7E260A:G oral microbiome included increased landscape complexity indicative of dysbiosis, and a significant increase of some bacteria, particularly Staphylococcus. These results suggest that normal alpha7 function plays a relevant role in maintaining normal gene expression and oral microbiome stasis. Consequently, this mouse model suggests there are consequences to ongoing alpha7 receptor dysfunction and oral health, as can occur from chronic exposure to nicotine as expected from electronic nicotine delivery systems (ENDS or "vaping"), that may not be seen until older age.
Collapse
Affiliation(s)
- Scott W Rogers
- Salt Lake City Veterans Affairs Medical Center: Geriatrics, Research, Education and Clinical Center, Salt Lake City, UT, United States.,Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Elizabeth J Myers
- Department of Internal Medicine, Division of Geriatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Lorise C Gahring
- Salt Lake City Veterans Affairs Medical Center: Geriatrics, Research, Education and Clinical Center, Salt Lake City, UT, United States.,Department of Internal Medicine, Division of Geriatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
16
|
Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, Gyengesi E. Cholinergic Modulation of Glial Function During Aging and Chronic Neuroinflammation. Front Cell Neurosci 2020; 14:577912. [PMID: 33192323 PMCID: PMC7594524 DOI: 10.3389/fncel.2020.577912] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex biological process that increases the risk of age-related cognitive degenerative diseases such as dementia, including Alzheimer’s disease (AD), Lewy Body Dementia (LBD), and mild cognitive impairment (MCI). Even non-pathological aging of the brain can involve chronic oxidative and inflammatory stress, which disrupts the communication and balance between the brain and the immune system. There has been an increasingly strong connection found between chronic neuroinflammation and impaired memory, especially in AD. While microglia and astrocytes, the resident immune cells of the central nervous system (CNS), exerting beneficial effects during the acute inflammatory phase, during chronic neuroinflammation they can become more detrimental. Central cholinergic circuits are involved in maintaining normal cognitive function and regulating signaling within the entire cerebral cortex. While neuronal-glial cholinergic signaling is anti-inflammatory and anti-oxidative, central cholinergic neuronal degeneration is implicated in impaired learning, memory sleep regulation, and attention. Although there is evidence of cholinergic involvement in memory, fewer studies have linked the cholinergic anti-inflammatory and anti-oxidant pathways to memory processes during development, normal aging, and disease states. This review will summarize the current knowledge of cholinergic effects on microglia and astroglia, and their role in both anti-inflammatory and anti-oxidant mechanisms, concerning normal aging and chronic neuroinflammation. We provided details on how stimulation of α7 nicotinic acetylcholine (α7nACh) receptors can be neuroprotective by increasing amyloid-β phagocytosis, decreasing inflammation and reducing oxidative stress by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and decreasing the release of pro-inflammatory cytokines. There is also evidence for astroglial α7nACh receptor stimulation mediating anti-inflammatory and antioxidant effects by inhibiting the nuclear factor-κB (NF-κB) pathway and activating the Nrf2 pathway respectively. We conclude that targeting cholinergic glial interactions between neurons and glial cells via α7nACh receptors could regulate neuroinflammation and oxidative stress, relevant to the treatment of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rashmi Gamage
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ingrid Wagnon
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ilaria Rossetti
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ryan Childs
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
17
|
Pinheiro L, Faustino C. Therapeutic Strategies Targeting Amyloid-β in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:418-452. [PMID: 30907320 DOI: 10.2174/1567205016666190321163438] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein misfolding and aggregation. AD is pathologically characterized by senile plaques formed by extracellular Amyloid-β (Aβ) peptide and Intracellular Neurofibrillary Tangles (NFT) formed by hyperphosphorylated tau protein. Extensive synaptic loss and neuronal degeneration are responsible for memory impairment, cognitive decline and behavioral dysfunctions typical of AD. Amyloidosis has been implicated in the depression of acetylcholine synthesis and release, overactivation of N-methyl-D-aspartate (NMDA) receptors and increased intracellular calcium levels that result in excitotoxic neuronal degeneration. Current drugs used in AD treatment are either cholinesterase inhibitors or NMDA receptor antagonists; however, they provide only symptomatic relief and do not alter the progression of the disease. Aβ is the product of Amyloid Precursor Protein (APP) processing after successive cleavage by β- and γ-secretases while APP proteolysis by α-secretase results in non-amyloidogenic products. According to the amyloid cascade hypothesis, Aβ dyshomeostasis results in the accumulation and aggregation of Aβ into soluble oligomers and insoluble fibrils. The former are synaptotoxic and can induce tau hyperphosphorylation while the latter deposit in senile plaques and elicit proinflammatory responses, contributing to oxidative stress, neuronal degeneration and neuroinflammation. Aβ-protein-targeted therapeutic strategies are thus a promising disease-modifying approach for the treatment and prevention of AD. This review summarizes recent findings on Aβ-protein targeted AD drugs, including β-secretase inhibitors, γ-secretase inhibitors and modulators, α-secretase activators, direct inhibitors of Aβ aggregation and immunotherapy targeting Aβ, focusing mainly on those currently under clinical trials.
Collapse
Affiliation(s)
- Lídia Pinheiro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| | - Célia Faustino
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| |
Collapse
|
18
|
Stanchfield ML, Webster SE, Webster MK, Linn CL. Involvement of HB-EGF/Ascl1/Lin28a Genes in Dedifferentiation of Adult Mammalian Müller Glia. Front Mol Biosci 2020; 7:200. [PMID: 32923455 PMCID: PMC7457012 DOI: 10.3389/fmolb.2020.00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Previous studies from this lab have determined that dedifferentiation of Müller glia occurs after eye drop application of an α7 nicotinic acetylcholine receptor (nAChR) agonist, PNU-282987, to the adult rodent eye. PNU-282987 acts on α7 nAChRs on retinal pigment epithelial cells to stimulate production of Müller-derived progenitor cells (MDPCs) and ultimately lead to neurogenesis. This current study was designed to test the hypothesis that the activation of genes involved in the HB-EGF/Ascl1/Lin28a signaling pathway in Müller glia leads to the genesis of MDPCs. RNA-seq was performed on a Müller glial cell line (rMC-1) following contact with supernatant collected from a retinal pigment epithelial (RPE) cell line treated with PNU-282987. Differentially regulated genes were compared with published literature of Müller glia dedifferentiation that occurs in lower vertebrate regeneration and early mammalian development. HB-EGF was significantly up-regulated by 8 h and expression increased through 12 h. By 48 h, up-regulation of Ascl1 and Lin28a was observed, two genes known to be rapidly induced in dedifferentiating zebrafish Müller glia. Up-regulation of other genes known to be involved in mammalian development and zebrafish regeneration were also observed, as well as down-regulation of some factors necessary for Müller glia cell identity. RNA-seq results were verified using qRT-PCR. Using immunocytochemistry, the presence of markers associated with MDCP identity, Otx2, Nestin, and Vsx2, were found to be expressed in the 48 h treatment group cultures. This study is novel in its demonstration that Müller glia in adult rodents can be induced into regenerative activity by stimulating genes involved in the HB-EGF/Ascl1/Lin28a pathway that leads to MDPCs after introducing conditioned media from PNU-282987 treated RPE. This study furthers our understanding of the mechanism by which Müller glia dedifferentiate in response to PNU-282987 in the adult mammalian retina.
Collapse
Affiliation(s)
| | | | | | - Cindy L. Linn
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, United States
| |
Collapse
|
19
|
Zirak MR, Karimi G, Rahimian R, Jafarian AH, Hayes AW, Mehri S. Tropisetron ameliorates cyclophosphamide-induced hemorrhagic cystitis in rats. Eur J Pharmacol 2020; 883:173310. [PMID: 32619674 DOI: 10.1016/j.ejphar.2020.173310] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023]
Abstract
Hemorrhagic cystitis is one of the most important complications of cyclophosphamide, a drug widely used in cancer chemotherapy and bone marrow transplantation. 5-HT3 antagonists are anti-emetic agents and have been shown to have notable anti-inflammatory and antioxidant properties. This study was designed to investigate the possible protective effects of tropisetron against cyclophosphamide-induced hemorrhagic cystitis in rats. Hemorrhagic cystitis was induced in female rats by cyclophosphamide (270 mg/kg). Tropisetron (2.5, 5 and 7.5 mg/kg), granisetron (2.5 and 5 mg/kg), and ondansetron (5 mg/kg) were injected 15 min before, 4 and 8 h after cyclophosphamide. To evaluate the role of alpha7 nicotinic acetylcholine receptor (α7nAChR), its antagonist, methyllycaconitine (5 mg/kg) was administered 30 min before tropisetron. After 24 h, animals were killed under anesthesia. Macroscopic and histological changes were evaluated. Malondialdehyde (MDA), glutathione (GSH) and Evans blue were measured spectrophotometrically. Furthermore, the protein levels of p38 mitogen-activated protein kinases (P38 MAPK), p-P38, signal transducer and activator of transcription 3 (STAT3), p-STAT3 and Poly (ADP-ribose) polymerase (PARP) were determined using Western blot. Cyclophosphamide administration significantly induced histopathological damages and increased MDA, p-p38/p38, p-STAT3/STAT3, and PARP levels compared with the saline group. Tropisetron treatment diminished histopathological injuries as well as MDA level, and STAT3 activity compared to cyclophosphamide treated rats. Co-administration of methyllycaconitine with tropisetron, partially or completely reversed the protective effects of tropisetron. Our results showed that prophylactic administration of tropisetron markedly ameliorated the cyclophosphamide-induced bladder hemorrhage and inflammation in rats. These effects of tropisetron were α7nAChR dependent.
Collapse
Affiliation(s)
- Mohammad Reza Zirak
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Amir Hossein Jafarian
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA; Michigan State University, East Lansing, MI, USA
| | - Soghra Mehri
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Zhu Z, Zhang L, Cui Y, Li M, Ren R, Li G, Sun X, Li Q. Functional Compensation and Mechanism of Choline Acetyltransferase in the Treatment of Cognitive Deficits in Aged Dementia Mice. Neuroscience 2020; 442:41-53. [PMID: 32497760 DOI: 10.1016/j.neuroscience.2020.05.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022]
Abstract
Choline acetyltransferase (ChAT) synthesizes the neurotransmitter acetylcholine (Ach). Exogenous supplementation with ChAT can functionally compensate for decreased Ach levels and ameliorate memory and cognitive deficits. In this paper, the treatment efficacy of recombinant ChAT (peptide transduction domain (PTD)-ChAT) and donepezil were compared in aged dementia mice, and their mechanisms were explored by performing the gene function annotation and enrichment analysis of differentially expressed genes. The Morris water maze test showed that the swimming times of PTD-ChAT-treated (4 mg/kg) and donepezil-treated (0.5 mg/kg) mice with mild and moderate dementia were significantly shortened (P < 0.01 vs aged dementia mice), and no significant changes were observed between the PTD-ChAT- and donepezil-treated groups. In contrast, the swimming times of PTD-ChAT-treated mice with severe dementia were noticeably shorter than those of donepezil-treated mice with severe dementia (P < 0.01), indicating that the treatment efficacy of PTD-ChAT is superior to that of donepezil. The effect of PTD-ChAT was further confirmed in transgenic dementia mice (C57BL/6J-TgN (APP/PS1) ZLFILAS). Gene function annotation and enrichment analysis showed that PTD-ChAT improved cognitive deficits through Ach and was implicated in neuroprotection, synaptic plasticity, neuronal survival, and cerebrovascular remodeling through ACh and vascular endothelial growth factor (VEGF) pathway activation. Donepezil was significantly correlated with the immune inflammatory response and the insulin and IGF-1 signaling pathways. Therefore, although PTD-ChAT and donepezil were both effective in the treatment of aged dementia mice, their mechanisms were significantly different. Our research indicated that PTD-ChAT has potential promise for research on new drugs for AD treatment.
Collapse
Affiliation(s)
- Zhenxia Zhu
- Capital Medical University Electric Power Teaching Hospital, No 1 Taipingqiaoxili, Beijing 100073, China
| | - Lulu Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Yali Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Meng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Rutong Ren
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Guoxing Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Xin Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Qian Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| |
Collapse
|
21
|
Oliveros-Matus P, Perez-Urrutia N, Alvarez-Ricartes N, Echeverria F, Barreto GE, Elliott J, Iarkov A, Echeverria V. Cotinine Enhances Fear Extinction and Astrocyte Survival by Mechanisms Involving the Nicotinic Acetylcholine Receptors Signaling. Front Pharmacol 2020; 11:303. [PMID: 32300297 PMCID: PMC7142247 DOI: 10.3389/fphar.2020.00303] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/28/2020] [Indexed: 01/03/2023] Open
Abstract
Fear memory extinction (FE) is an important therapeutic goal for Posttraumatic stress disorder (PTSD). Cotinine facilitates FE in rodents, in part due to its inhibitory effect on the amygdala by the glutamatergic projections from the medial prefrontal cortex (mPFC). The cellular and behavioral effects of infusing cotinine into the mPFC on FE, astroglia survival, and the expression of bone morphogenetic proteins (BMP) 2 and 8, were assessed in C57BL/6 conditioned male mice. The role of the α4β2- and α7 nicotinic acetylcholine receptors (nAChRs) on cotinine’s actions were also investigated. Cotinine infused into the mPFC enhanced contextual FE and decreased BMP8 expression by a mechanism dependent on the α7nAChRs. In addition, cotinine increased BMP2 expression and prevented the loss of GFAP + astrocytes in a form independent on the α7nAChRs but dependent on the α4β2 nAChRs. This evidence suggests that cotinine exerts its effect on FE by modulating nAChRs signaling in the brain.
Collapse
Affiliation(s)
- Patricia Oliveros-Matus
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Nelson Perez-Urrutia
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Nathalie Alvarez-Ricartes
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Florencia Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - James Elliott
- Northern Sydney Local Health District, The Kolling Research Institute and Faculty of Health Sciences, The University of Sydney, St. Leonards, NSW, Australia.,Department of Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alexandre Iarkov
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Valentina Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile.,Research and Development Department, Bay Pines VA Healthcare System, Bay Pines, FL, United States
| |
Collapse
|
22
|
Lykhmus O, Kalashnyk O, Uspenska K, Skok M. Positive Allosteric Modulation of Alpha7 Nicotinic Acetylcholine Receptors Transiently Improves Memory but Aggravates Inflammation in LPS-Treated Mice. Front Aging Neurosci 2020; 11:359. [PMID: 31998114 PMCID: PMC6966166 DOI: 10.3389/fnagi.2019.00359] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation accompanies or even precedes the development of cognitive changes in many brain pathologies, including Alzheimer’s disease. Therefore, dampening inflammatory reactions within the brain is a promising strategy for supporting cognitive functions in elderly people and for preventing the development of neurodegenerative disorders. Nicotinic acetylcholine receptors containing α7 subunits (α7 nAChRs) are involved in regulating cell survival, inflammation, and memory. The aim of our study was to evaluate the efficiency of α7-specific therapy at different stages of inflammation and to compare the effects of orthosteric agonist PNU282987 and type 2 positive allosteric modulator (PAM) PNU120596 in mice after a single injection of lipopolysaccharide (LPS). The data presented demonstrate that PNU282987 protected mice from LPS-induced impairment of episodic memory by decreasing IL-6 levels in the blood, stabilizing the brain mitochondria and up-regulating the brain α7-, α3-, and α4-containing nAChRs. Such treatment was efficient when given simultaneously with LPS or a week after LPS injection and was not efficient if LPS had been injected 2 months before. PNU120596 also decreased IL-6, stabilized mitochondria and up-regulated the brain nAChRs. However, its memory-improving effect was transient and disappeared after the end of the injection cycle. Moreover, cessation of PNU120596 treatment resulted in a sharp increase in IL-1β and IL-6 levels in the blood. It is concluded that activating α7 nAChRs protects the mouse brain from the pathogenic effect of LPS in the early stages of inflammation but is not efficient when irreversible changes have already occurred. The use of a PAM does not improve the effect of the agonist, possibly potentiates the effect of endogenous agonists, and results in undesirable effects after treatment cessation.
Collapse
Affiliation(s)
- Olena Lykhmus
- Immunology of Cellular Receptors, Department of Molecular Immunology, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Olena Kalashnyk
- Immunology of Cellular Receptors, Department of Molecular Immunology, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Kateryna Uspenska
- Immunology of Cellular Receptors, Department of Molecular Immunology, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Maryna Skok
- Immunology of Cellular Receptors, Department of Molecular Immunology, Palladin Institute of Biochemistry, Kyiv, Ukraine
| |
Collapse
|
23
|
Al Rawashdah S, Hamrouni A, Sadek B, Amer R, Metwaly M, Atatreh N, Ghattas MA. Molecular modelling studies on ɑ7 nicotinic receptor allosteric modulators yields novel filter-based virtual screening protocol. J Mol Graph Model 2019; 92:44-54. [DOI: 10.1016/j.jmgm.2019.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 11/26/2022]
|
24
|
Nicoletti CG, Landi D, Monteleone F, Mataluni G, Albanese M, Lauretti B, Rocchi C, Simonelli I, Boffa L, Buttari F, Mercuri NB, Centonze D, Marfia GA. Treatment with Dimethyl Fumarate Enhances Cholinergic Transmission in Multiple Sclerosis. CNS Drugs 2019; 33:1133-1139. [PMID: 31650471 DOI: 10.1007/s40263-019-00676-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) exerts anti-inflammatory effects in multiple sclerosis by activating the Nrf2 antioxidant pathway, which is also stimulated by acetylcholine via alpha-7 nicotinic acetylcholine receptors. In animal models, Nrf2 potentiates cholinergic synaptic plasticity. OBJECTIVE The aim of this study was to test whether treatment with DMF modulates cholinergic pathways in relapsing-remitting multiple sclerosis (RRMS). METHODS Patients starting DMF (20) or IFN-β 1a (20) and healthy subjects (20) were enrolled. Short-latency afferent inhibition (SAI), which is a transcranial stimulation measure of central cholinergic transmission, was recorded in patients and controls at baseline and, in patients only, after 6 months of treatment. Patients treated with DMF also underwent autonomic function testing to further explore peripheral and central cholinergic tone. RESULTS At baseline, SAI was similar in patients and in controls (p = 0.983). Treatment with DMF significantly increased SAI (p = 0.01), while IFNβ had no effect (p = 0.80). In the cold face test, DMF treatment also increased reflex bradycardia (p = 0.013), and reduced diastolic blood pressure variation (p = 0.010), further indicating its ability to stimulate cholinergic transmission. CONCLUSIONS Treatment of MS patients with DMF results in increased cholinergic stimulation, with possible implications for neuroinflammation and neuroprotection.
Collapse
Affiliation(s)
- Carolina Gabri Nicoletti
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Doriana Landi
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Fabrizia Monteleone
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Giorgia Mataluni
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Maria Albanese
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Benedetta Lauretti
- Neurology Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Camilla Rocchi
- Neurology Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Ilaria Simonelli
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy.,Service of Medical Statistics and Information Technology, Fondazione Fatebenefratelli per la Ricerca e la Formazione Sanitaria e Sociale, Lungotevere de' Cenci 5, 00186, Rome, Italy
| | - Laura Boffa
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy
| | - Fabio Buttari
- Neurology and Neurorehabilitation Units, IRCCS NEUROMED, Via Atinense 18, 86077, Pozzilli, IS, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy.,Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Diego Centonze
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy. .,Neurology and Neurorehabilitation Units, IRCCS NEUROMED, Via Atinense 18, 86077, Pozzilli, IS, Italy.
| | - Girolama Alessandra Marfia
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University and Hospital, Via Montpellier 1, 00133, Rome, Italy.,Neurology and Neurorehabilitation Units, IRCCS NEUROMED, Via Atinense 18, 86077, Pozzilli, IS, Italy
| |
Collapse
|
25
|
Pérez de Vega MJ, Fernandez-Mendivil C, de la Torre Martínez R, González-Rodríguez S, Mullet J, Sala F, Sala S, Criado M, Moreno-Fernández S, Miguel M, Fernández-Carvajal A, Ferrer-Montiel A, López MG, González-Muñiz R. 1-(2',5'-Dihydroxyphenyl)-3-(2-fluoro-4-hydroxyphenyl)-1-propanone (RGM079): A Positive Allosteric Modulator of α7 Nicotinic Receptors with Analgesic and Neuroprotective Activity. ACS Chem Neurosci 2019; 10:3900-3909. [PMID: 31322853 DOI: 10.1021/acschemneuro.9b00364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acetylcholine α7 nicotinic receptors are widely expressed in the brain, where they are involved in the central processing of pain as well as in neuropsychiatric, neurodegenerative, and inflammatory processes. Positive allosteric modulators (PAMs) show the advantage of allowing the selective regulation of different subtypes of acetylcholine receptors without directly interacting with the agonist binding site. Here, we report the preparation and biological activity of a fluoro-containing compound, 1-(2',5'-dihydroxyphenyl)-3-(2-fluoro-4-hydroxyphenyl)-1-propanone (8, RGM079), that behaves as a potent PAM of the α7 receptors and has a balanced pharmacokinetic profile and antioxidant properties comparable or even higher than well-known natural polyphenols. In addition, compound RGM079 shows neuroprotective properties in Alzheimer's disease (AD)-toxicity related models. Thus, it causes a concentration-dependent neuroprotective effect against the toxicity induced by okadaic acid (OA) in the human neuroblastoma cell line SH-SY5Y. Similarly, in primary cultures of rat cortical neurons, RGM079 is able to restore the cellular viability after exposure to OA and amyloid peptide Aβ1-42, with cell death almost completely prevented at 10 and 30 μM, respectively. Finally, compound RGM079 shows in vivo analgesic activity in the complete Freund's adjuvant (CFA)-induced paw inflammation model after intraperitoneal administration.
Collapse
Affiliation(s)
| | - Cristina Fernandez-Mendivil
- Instituto Teófilo Hernando, Department of Pharmacology, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, Madrid 28029, Spain
| | - Roberto de la Torre Martínez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avenida de la Universidad s/n, Elche, Alicante 03202, Spain
| | - Sara González-Rodríguez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avenida de la Universidad s/n, Elche, Alicante 03202, Spain
| | - José Mullet
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Alicante 03500, Spain
| | - Francisco Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Alicante 03500, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Alicante 03500, Spain
| | - Manuel Criado
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Alicante 03500, Spain
| | - Silvia Moreno-Fernández
- Instituto de Investigación en Ciencias de la Alimentación (CSIC-UAM), C/Nicolás Cabrera 9, Madrid 28049, Spain
| | - Marta Miguel
- Instituto de Investigación en Ciencias de la Alimentación (CSIC-UAM), C/Nicolás Cabrera 9, Madrid 28049, Spain
| | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avenida de la Universidad s/n, Elche, Alicante 03202, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avenida de la Universidad s/n, Elche, Alicante 03202, Spain
| | - Manuela G. López
- Instituto Teófilo Hernando, Department of Pharmacology, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, Madrid 28029, Spain
| | | |
Collapse
|
26
|
Yao C, Liu X, Zhou Z, Xiang Y, Yuan S, Xie W, Zhou M, Hu Z, Li Y, Ji A, Cai T. Melatonin attenuates expression of cyclooxygenase-2 (COX-2) in activated microglia induced by lipopolysaccharide (LPS). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:437-446. [PMID: 31081481 DOI: 10.1080/15287394.2019.1615019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Lipopolysaccharide (LPS) is a known neurotoxin and utilized most extensively as a microglial activator for induction of inflammatory neurodegeneration. Melatonin (MEL) is the main secretory product of pineal gland reported to be responsible for a variety of physiological functions. However, the molecular mechanisms underlying the influence of MEL on microglia activation remain unclear. The aim of this study was to investigate the effect of MEL on cyclooxygenase-2 (COX-2) levels in LPS-induced microglia. The results of RT-PCR and Western blot analysis showed that MEL significantly inhibited LPS-mediated upregulation of COX-2 in microglia. Data from ELISA demonstrated that prostaglandin E2 (PGE2), the downstream effector of COX-2, concentrations were also reduced. In addition, MEL was found to decrease activation of ERK1/2, JNK, p38 MAPK, and NF-κB, the upstream signal pathways of COX-2. Taken together, evidence indicates that MEL may attenuate upregulation of COX-2 by blocking the MAPK/NF-κB signaling pathway in LPS-stimulated microglia.
Collapse
Affiliation(s)
- Chunyan Yao
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Xiaoling Liu
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Zhengyu Zhou
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Ying Xiang
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Shuai Yuan
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Weijia Xie
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Meiyu Zhou
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Zeyao Hu
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Yafei Li
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| | - Ailing Ji
- b Department of Preventive Medicine & Chongqing Engineering Research Center of Pharmaceutical Sciences , Chongqing Medical and Pharmaceutical College , Chongqing , China
| | - Tongjian Cai
- a Department of Epidemiology , College of Preventive Medicine, Army Medical University (Third Military Medical University) , Chongqing , China
| |
Collapse
|
27
|
Manetti D, Garifulina A, Bartolucci G, Bazzicalupi C, Bellucci C, Chiaramonte N, Dei S, Di Cesare Mannelli L, Ghelardini C, Gratteri P, Spirova E, Shelukhina I, Teodori E, Varani K, Tsetlin V, Romanelli MN. New Rigid Nicotine Analogues, Carrying a Norbornane Moiety, Are Potent Agonists of α7 and α3* Nicotinic Receptors. J Med Chem 2019; 62:1887-1901. [PMID: 30681854 DOI: 10.1021/acs.jmedchem.8b01372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A three-dimensional database search has been applied to design a series of endo- and exo-3-(pyridin-3-yl)bicyclo[2.2.1]heptan-2-amines as nicotinic receptor ligands. The synthesized compounds were tested in radioligand binding assay on rat cortex against [3H]-cytisine and [3H]-methyllycaconitine to measure their affinity for α4β2* and α7* nicotinic receptors. The new derivatives showed some preference for the α4β2* over the α7* subtype, with their affinity being dependent on the endo/exo isomerism and on the methylation degree of the basic nitrogen. The endo primary amines displayed the lowest Ki values on both receptor subtypes. Selected compounds (1a, 2a, 3a, and 6a) were tested on heterologously expressed α4β2, α7, and α3β2 receptors and on SHSY-5Y cells. Compounds 1a and 2a showed α4β2 antagonistic properties while behaved as full agonists on recombinant α7 and on SHSY5Y cells. On the α3β2 subtype, only the chloro derivative 2a showed full agonist activity and submicromolar potency (EC50 = 0.43 μM). The primary amines described here represent new chemotypes for the α7 and α3* receptor subtypes.
Collapse
Affiliation(s)
- Dina Manetti
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Alexandra Garifulina
- Department of Molecular Basis of Neurosignaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Russian Academy of Sciences , Miklukho-Maklaya Street 16/10 , 117997 Moscow , Russia
| | - Gianluca Bartolucci
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Carla Bazzicalupi
- Department of Chemistry "Ugo Schiff" , University of Florence , Via della Lastruccia 3 , 50019 Sesto Fiorentino , Italy
| | - Cristina Bellucci
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Niccolò Chiaramonte
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Silvia Dei
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Lorenzo Di Cesare Mannelli
- Department of NEUROFARBA, Section of Pharmacology and Toxicology , University of Florence , Viale G Pieraccini 6 , 50139 Firenze , Italy
| | - Carla Ghelardini
- Department of NEUROFARBA, Section of Pharmacology and Toxicology , University of Florence , Viale G Pieraccini 6 , 50139 Firenze , Italy
| | - Paola Gratteri
- Department of NEUROFARBA, Laboratory of Molecular Modeling Cheminformatics & QSAR , University of Firenze , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Ekaterina Spirova
- Department of Molecular Basis of Neurosignaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Russian Academy of Sciences , Miklukho-Maklaya Street 16/10 , 117997 Moscow , Russia
| | - Irina Shelukhina
- Department of Molecular Basis of Neurosignaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Russian Academy of Sciences , Miklukho-Maklaya Street 16/10 , 117997 Moscow , Russia
| | - Elisabetta Teodori
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| | - Katia Varani
- Institute of Pharmacology , University of Ferrara , Via Fossato di Mortara 17-19 , 44100 Ferrara , Italy
| | - Victor Tsetlin
- Department of Molecular Basis of Neurosignaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Russian Academy of Sciences , Miklukho-Maklaya Street 16/10 , 117997 Moscow , Russia
| | - Maria Novella Romanelli
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Via Ugo Schiff 6 , 50019 Sesto Fiorentino , Italy
| |
Collapse
|
28
|
Spirova EN, Ivanov IA, Kasheverov IE, Kudryavtsev DS, Shelukhina IV, Garifulina AI, Son LV, Lummis SCR, Malca-Garcia GR, Bussmann RW, Hennig L, Giannis A, Tsetlin VI. Curare alkaloids from Matis Dart Poison: Comparison with d-tubocurarine in interactions with nicotinic, 5-HT3 serotonin and GABAA receptors. PLoS One 2019; 14:e0210182. [PMID: 30608952 PMCID: PMC6319706 DOI: 10.1371/journal.pone.0210182] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Several novel bisbenzylisoquinoline alkaloids (BBIQAs) have recently been isolated from a Matis tribe arrow poison and shown by two-electrode voltage-clamp to inhibit mouse muscle nicotinic acetylcholine receptors (nAChR). Here, using radioligand assay with Aplysia californica AChBP and radioiodinated α-bungarotoxin ([125I]-αBgt), we show that BBIQA1, BBIQA2, and d-tubocurarine (d-TC) have similar affinities to nAChR orthosteric site. However, a competition with [125I]-αBgt for binding to the Torpedo californica muscle-type nAChR revealed that BBIQAs1, 2, and 3 are less potent (IC50s = 26.3, 8.75, and 17.0 μM) than d-TC (IC50 = 0.39 μM), while with α7 nAChR in GH4C1 cells, BBIQA1 was less potent that d-TC (IC50s = 162 μM and 7.77 μM, respectively), but BBIQA2 was similar (IC50 = 5.52 μM). In inhibiting the Ca2+ responses induced by acetylcholine in Neuro2a cells expressing the mouse adult α1β1εδ nAChR or human α7 nAChR, BBIQAs1 and 2 had similar potencies to d-TC (IC50s in the range 0.75-3.08 μM). Our data suggest that BBIQA1 and BBIQA2 can inhibit adult muscle α1β1εδ nAChR by both competitive and noncompetitive mechanisms. Further experiments on neuronal α3β2, α4β2, and α9α10 nAChRs, expressed in Xenopus laevis oocytes, showed that similar potencies for BBIQAs1, 2, and d-TC. With α3β2γ2 GABAAR currents were almost completely inhibited by d-TC at a high (100 μM) concentration, but BBIQAs1 and 2 were less potent (only 40-50% inhibition), whereas in competition with Alexa Fluor 546-α-cobratoxin for binding to α1β3γ2 GABAAR in Neuro2a cells, d-TC and these analogs had comparable affinities. Especially interesting effects of BBIQAs1 and 2 in comparison with d-TC were observed for 5-HT3AR: BBIQA1 and BBIQA2 were 5- and 87-fold less potent than d-TC (IC50 = 22.63 nM). Thus, our results reveal that these BBIQAs differ from d-TC in their potencies towards certain Cys-loop receptors, and we suggest that understanding the reasons behind this might be useful for future drug design.
Collapse
Affiliation(s)
- Ekaterina N. Spirova
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Igor A. Ivanov
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Igor E. Kasheverov
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, Russia
| | - Denis S. Kudryavtsev
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Irina V. Shelukhina
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra I. Garifulina
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Lina V. Son
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Gonzalo R. Malca-Garcia
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, United States of America
| | | | - Lothar Hennig
- Institut für Organische Chemie, Fakultät für Chemie und Mineralogie, Universität Leipzig, Leipzig, Germany
| | - Athanassios Giannis
- Institut für Organische Chemie, Fakultät für Chemie und Mineralogie, Universität Leipzig, Leipzig, Germany
| | - Victor I. Tsetlin
- Department of Molecular Neuroimmune signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- PhysBio of MEPhI, Moscow, Russia
| |
Collapse
|
29
|
Effect of overnight smoking abstinence on a marker for microglial activation: a [ 11C]DAA1106 positron emission tomography study. Psychopharmacology (Berl) 2018; 235:3525-3534. [PMID: 30343364 PMCID: PMC6497451 DOI: 10.1007/s00213-018-5077-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/10/2018] [Indexed: 12/27/2022]
Abstract
RATIONALE Microglia are the main immune cells in the central nervous system and participate in neuroinflammation. When activated, microglia express increased levels of the translocator protein 18 kDa (TSPO), thereby making TSPO availability a marker for neuroinflammation. Using positron emission tomography (PET) scanning, our group recently demonstrated that smokers in the satiated state had 16.8% less binding of the radiotracer [11C]DAA1106 (a radioligand for TSPO) in the brain than nonsmokers. OBJECTIVES We sought to determine the effect of overnight smoking abstinence on [11C]DAA1106 binding in the brain. METHODS Forty participants (22 smokers and 18 nonsmokers) completed the study (at one of two sites) and had usable data, which included images from a dynamic [11C]DAA1106 PET scanning session (with smokers having been abstinent for 17.9 ± 2.3 h) and a blood sample for TSPO genotyping. Whole brain standardized uptake values (SUVs) were determined, and analysis of variance was performed, with group (overnight abstinent smoker vs. nonsmoker), site, and TSPO genotype as factors, thereby controlling for site and genotype. RESULTS Overnight abstinent smokers had lower whole brain SUVs (by 15.5 and 17.0% for the two study sites) than nonsmokers (ANCOVA, P = 0.004). The groups did not significantly differ in injected radiotracer dose or body weight, which were used to calculate SUV. CONCLUSIONS These results in overnight abstinent smokers are similar to those in satiated smokers, indicating that chronic cigarette smoking leads to global impairment of microglial activation which persists into early abstinence. Other explanations for study results, such as smoking leading to reduced numbers of microglia or smokers having more rapid metabolism of the radiotracer than nonsmokers, are also possible.
Collapse
|
30
|
Mulcahy MJ, Paulo JA, Hawrot E. Proteomic Investigation of Murine Neuronal α7-Nicotinic Acetylcholine Receptor Interacting Proteins. J Proteome Res 2018; 17:3959-3975. [PMID: 30285449 DOI: 10.1021/acs.jproteome.8b00618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.
Collapse
Affiliation(s)
- Matthew J Mulcahy
- Division of Biology and Biological Engineering , California Institute of Technology , 1200 East California Boulevard , Pasadena , California 91125-2900 , United States.,Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Edward Hawrot
- Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
31
|
Cholinergic Overstimulation Attenuates Rule Selectivity in Macaque Prefrontal Cortex. J Neurosci 2017; 38:1137-1150. [PMID: 29255006 DOI: 10.1523/jneurosci.3198-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 11/21/2022] Open
Abstract
Acetylcholine is released in the prefrontal cortex (PFC) and is a key modulator of cognitive performance in primates. Cholinergic stimulation has been shown to have beneficial effects on performance of cognitive tasks, and cholinergic receptors are being actively explored as promising targets for ameliorating cognitive deficits in Alzheimer's disease. We hypothesized that cholinergic stimulation of PFC during performance of a cognitive task would augment neuronal activity and neuronal coding of task attributes. We iontophoretically applied the general cholinergic receptor agonist carbachol onto neurons in dorsolateral PFC (DLPFC) of male rhesus macaques performing rule-guided prosaccades and antisaccades, a well established oculomotor task for testing cognitive control. Carbachol application had heterogeneous effects on neuronal excitability, with both excitation and suppression observed in significant proportions. Contrary to our prediction, neurons with rule-selective activity exhibited a reduction in selectivity during carbachol application. Cholinergic stimulation disrupted rule selectivity regardless of whether it had suppressive or excitatory effects on these neurons. In addition, cholinergic stimulation excited putative pyramidal neurons, whereas the activity of putative interneurons remained unchanged. Moreover, cholinergic stimulation attenuated saccade direction selectivity in putative pyramidal neurons due to nonspecific increases in activity. Our results suggest excessive cholinergic stimulation has detrimental effects on DLPFC representations of task attributes. These findings delineate the complexity and heterogeneity of neuromodulation of cerebral cortex by cholinergic stimulation, an area of active exploration with respect to the development of cognitive enhancers.SIGNIFICANCE STATEMENT The neurotransmitter acetylcholine is known to be important for cognitive processes in the prefrontal cortex. Removal of acetylcholine from prefrontal cortex can disrupt short-term memory performance and is reminiscent of Alzheimer's disease, which is characterized by degeneration of acetylcholine-producing neurons. Stimulation of cholinergic receptors is being explored to create cognitive enhancers for the treatment of Alzheimer's disease and other psychiatric diseases. Here, we stimulated cholinergic receptors in prefrontal cortex and examined its effects on neurons that are engaged in cognitive behavior. Surprisingly, cholinergic stimulation decreased neurons' ability to discriminate between rules. This work suggests that overstimulation of acetylcholine receptors could disrupt neuronal processing during cognition and is relevant to the design of cognitive enhancers based on stimulating the cholinergic system.
Collapse
|