1
|
Rosenthal JS, Zhang D, Yin J, Long C, Yang G, Li Y, Lu Z, Li WP, Yu Z, Li J, Yuan Q. Molecular organization of central cholinergic synapses. Proc Natl Acad Sci U S A 2025; 122:e2422173122. [PMID: 40273107 PMCID: PMC12054790 DOI: 10.1073/pnas.2422173122] [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: 10/29/2024] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Synapses have undergone significant diversification and adaptation, contributing to the complexity of the central nervous system. Understanding their molecular architecture is essential for deciphering the brain's functional evolution. While nicotinic acetylcholine receptors (nAchRs) are widely distributed across metazoan brains, their associated protein networks remain poorly characterized. Using in vivo proximity labeling, we generated proteomic maps of subunit-specific nAchR interactomes in developing and mature Drosophila brains. Our findings reveal a developmental expansion and reconfiguration of the nAchR interactome. Proteome profiling with genetic perturbations showed that removing individual nAchR subunits consistently triggers compensatory shifts in receptor subtypes, highlighting mechanisms of synaptic plasticity. We also identified the Rho-GTPase regulator Still life (Sif) as a key organizer of cholinergic synapses, with loss of Sif disrupting their molecular composition and structural integrity. These results provide molecular insights into the development and plasticity of central cholinergic synapses, advancing our understanding of synaptic identity conservation and divergence.
Collapse
Affiliation(s)
- Justin S. Rosenthal
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - Dean Zhang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - Jun Yin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - Caixia Long
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - George Yang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - Yan Li
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| | - Zhiyuan Lu
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Wei-Ping Li
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Zhiheng Yu
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Jiefu Li
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Quan Yuan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD20892
| |
Collapse
|
2
|
Ito R, Ojima H, Takebayashi M, Takeuchi M, Takahashi H, Mori S, Ihara M, Sattelle DB, Matsuda K. Binding site loops D and G make a stronger contribution than loop C to the actions of neonicotinoids on the NACHO-assisted, robustly expressed Drosophila melanogaster Dα1/Dβ1 nicotinic acetylcholine receptor. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 209:106325. [PMID: 40082022 DOI: 10.1016/j.pestbp.2025.106325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
TMX3 is essential for the functional expression of insect nicotinic acetylcholine receptors (nAChRs) and RIC-3 and UNC-50 modulate it. In addition to these cofactors, NACHO has been shown to enhance functional expression of certain vertebrate nAChRs and an insect homomeric nAChR. Here, we have examined the impact of Drosophila melanogaster NACHO (DmNACHO) on the ACh-induced response amplitude of the fruit fly Dα1/Dβ1 nAChRs coexpressed in Xenopus laevis oocytes with DmRIC-3, DmTMX3 and DmUNC-50 and examined the actions of neonicotinoid insecticides. DmNACHO markedly enhanced the ACh and neonicotinoid-induced response amplitude of Dα1/Dβ1 nAChRs coexpressed with the three cofactors DmRIC-3, DmTMX3 and DmUNC-50, while scarcely influencing ligand affinity. Given the robust Dα1/Dβ1 nAChR expression with the aid of the four cofactors, we investigated the impact of mutations in loops C, D and G of the orthosteric ligand binding domain (LBD) on the actions of the neonicotinoids imidacloprid and thiacloprid. Both the R81T mutation in loop D and the A60S mutation in loop G significantly reduced the agonist actions of the neonicotinoids, whereas the S221E mutation in loop C had no significant effect on agonist actions on this nAChR. Further, no greater affinity and efficacy reducing effects were observed even when the S221E mutation in loop C was combined with the R81T mutation in loop D, the A60S mutation in loop G, or both, demonstrating that loop D and loop G are more critical than loop C in determining the target-site actions of imidacloprid and thiacloprid.
Collapse
Affiliation(s)
- Ryo Ito
- Graduate School of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Hisanori Ojima
- Graduate School of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Mayuka Takebayashi
- Graduate School of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Miyu Takeuchi
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Hiyori Takahashi
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Sumito Mori
- Graduate School of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - Makoto Ihara
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan
| | - David B Sattelle
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, London WC1E 6JF, United Kingdom
| | - Kazuhiko Matsuda
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan; Agricultural Technology and Innovation Research Institute, Kindai University, 3327-204 Nakamachi, Nara 631-8505, Japan.
| |
Collapse
|
3
|
Holmgård DSG, Zhou L, Kristensen JL, Jensen AA. The Heterogeneous Kinetic Origins of the Binding Properties of Orthosteric Ligands at Heteromeric Nicotinic Acetylcholine Receptors. J Med Chem 2025; 68:6683-6697. [PMID: 40043102 DOI: 10.1021/acs.jmedchem.5c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
A plethora of agonists and competitive antagonists have been developed to explore the therapeutic potential in neuronal nicotinic acetylcholine receptors (nAChRs). Based on equilibrium and kinetic [3H]epibatidine binding studies, we report that the kinetic fingerprints of [3H]epibatidine at five heteromeric αβ nAChRs and of seven classical agonists at α4β2 and α3β4 nAChRs differ substantially. While this diversity depends on both the agonist and receptor subtype, the overall pattern of kinetic determinants emerging from this profiling is complex. The dramatically different binding kinetics displayed by two alkaloids and competitive antagonists, (+)-DHβE and (+)-cocculine, at the α4β2 nAChR further exemplify how dissimilar kinetics can underlie very comparable pharmacological properties exhibited by close structural analogs. Thus, our findings elucidate the heterogeneous kinetic basis for orthosteric ligand binding to αβ nAChRs and emphasize how the binding affinities, selectivity profiles, and structure-activity relationships of these ligands are rooted in their kinetic traits at the receptors.
Collapse
Affiliation(s)
- David S G Holmgård
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Unversitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Libin Zhou
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Unversitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Jesper L Kristensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Unversitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Unversitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
4
|
Heydary YH, Castro EM, Lotfipour S, Leslie FM. "Unraveling the role of CHRNA6, the neuronal α6 nicotinic acetylcholine receptor subunit". RECEPTORS (BASEL, SWITZERLAND) 2025; 4:1. [PMID: 40331132 PMCID: PMC12051391 DOI: 10.3390/receptors4010001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
The increased prevalence of electronic cigarettes, particularly among adolescents, has escalated concerns about nicotine addiction. Nicotine, a potent psychostimulant found in tobacco products, exerts its effects by interacting with nicotinic acetylcholine receptors (nAChRs) in the brain. Recent findings in both pre-clinical and clinical studies have enhanced our understanding of nAChRs, overcoming limitations of pharmacological tools that previously hindered their investigation. Of particular interest is the α6 subunit, whose expression peaks during adolescence, a critical period of brain development often marked by the initiation of substance use. Pre-clinical studies have linked α6-containing nAChRs (α6*nAChRs) to nicotine-induced locomotion, dopamine release, and self-administration behavior. Furthermore, clinical studies suggest an association between the α6 subunit and increased smoking behavior in humans. Specifically, a single nucleotide polymorphism in the 3' untranslated region of the CHRNA6 gene that encodes for this subunit is linked to smoking behavior and other substance use. A comprehensive understanding of this subunit's role in addiction is of high importance. This review aims to consolidate current knowledge regarding the α6 subunit's functions and implications in addiction and other disorders, with the hope of paving the way for future research and the development of targeted therapies to address this pressing public health concern.
Collapse
Affiliation(s)
- Yasamin Hajy Heydary
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Emily M. Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
- Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Frances M. Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
5
|
Hooda Y, Sente A, Judy RM, Smalinskaitė L, Peak-Chew SY, Naydenova K, Malinauskas T, Hardwick SW, Chirgadze DY, Aricescu AR, Hegde RS. Mechanism of NACHO-mediated assembly of pentameric ligand-gated ion channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620708. [PMID: 39553992 PMCID: PMC11565801 DOI: 10.1101/2024.10.28.620708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Pentameric ligand-gated ion channels (pLGICs) are cell surface receptors of crucial importance for animal physiology1-4. This diverse protein family mediates the ionotropic signals triggered by major neurotransmitters and includes γ-aminobutyric acid receptors (GABAARs) and acetylcholine receptors (nAChRs). Receptor function is fine-tuned by a myriad of endogenous and pharmacological modulators3. A functional pLGIC is built from five homologous, sometimes identical, subunits, each containing a β-scaffold extracellular domain (ECD), a four-helix transmembrane domain (TMD) and intracellular loops of variable length. Although considerable progress has been made in understanding pLGICs in structural and functional terms, the molecular mechanisms that enable their assembly at the endoplasmic reticulum (ER)5 in a vast range of potential subunit configurations6 remain unknown. Here, we identified candidate pLGICs assembly factors selectively associated with an unassembled GABAAR subunit. Focusing on one of the candidates, we determined the cryo-EM structure of an assembly intermediate containing two α1 subunits of GABAAR each bound to an ER-resident membrane protein NACHO. The structure showed how NACHO shields the principal (+) transmembrane interface of α1 subunits containing an immature extracellular conformation. Crosslinking and structure-prediction revealed an adjacent surface on NACHO for β2 subunit interactions to guide stepwise oligimerisation. Mutations of either subunit-interacting surface on NACHO also impaired the formation of homopentameric α7 nAChRs, pointing to a generic framework for pLGIC assembly. Our work provides the foundation for understanding the regulatory principles underlying pLGIC structural diversity.
Collapse
Affiliation(s)
- Yogesh Hooda
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
- Equal contribution
| | - Andrija Sente
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
- Equal contribution
| | - Ryan M. Judy
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
- Equal contribution
| | - Luka Smalinskaitė
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Sew-Yeu Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | | | - Tomas Malinauskas
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Steven W. Hardwick
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, United Kingdom
| | - Dimitri Y. Chirgadze
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, United Kingdom
| | - A. Radu Aricescu
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | | |
Collapse
|
6
|
Baxter L, Hopkins S, O'Connor KC, Pham MC, Nowak RJ, Monson NL, Blackburn K, Hibbs RE, Vernino S, Noviello CM. Fluorescence-detection size-exclusion chromatography specifically detects autoantibodies targeting the ganglionic acetylcholine receptor in patients with autoimmune autonomic ganglionopathy. J Neuroimmunol 2024; 396:578454. [PMID: 39277987 DOI: 10.1016/j.jneuroim.2024.578454] [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: 06/17/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Autoimmune autonomic ganglionopathy (AAG) is a rare disease wherein autoantibodies target the ganglionic acetylcholine receptor (gAChR). Current diagnosis in the United States depends upon clinical symptoms and positive autoantibody detection using a radioimmunoprecipitation assay (RIA). Here we offer a proof-of-principle study on an alternative method, fluorescence-detection size-exclusion-chromatography (FSEC). We show FSEC can detect autoantibodies against gAChR from patient sera but not healthy controls or samples from other autoimmune diseases. We compare FSEC to RIA and find good correlation. We discuss potential advantages of using FSEC as an alternative or as a first-step diagnostic prior to pursuing existing methodologies.
Collapse
Affiliation(s)
- Leah Baxter
- Department of Neurobiology, University of California San Diego, United States
| | - Steven Hopkins
- Department of Neurology, University of Texas Southwestern Medical Center, United States
| | - Kevin C O'Connor
- Department of Neurology, Yale University School of Medicine, United States; Department of Immunobiology, Yale University School of Medicine, United States
| | - Minh C Pham
- Department of Neurology, Yale University School of Medicine, United States
| | - Richard J Nowak
- Department of Immunobiology, Yale University School of Medicine, United States
| | - Nancy L Monson
- Department of Neurology, University of Texas Southwestern Medical Center, United States
| | - Kyle Blackburn
- Department of Neurology, University of Texas Southwestern Medical Center, United States
| | - Ryan E Hibbs
- Department of Neurobiology, University of California San Diego, United States; Department of Pharmacology, University of California San Diego, United States
| | - Steven Vernino
- Department of Neurology, University of Texas Southwestern Medical Center, United States
| | - Colleen M Noviello
- Department of Neurobiology, University of California San Diego, United States.
| |
Collapse
|
7
|
Goebel S, Cordova-Martinez D, Verselis VK, Francesconi A. Dampened α7 nAChR activity contributes to audiogenic seizures and hyperactivity in a mouse model of Fragile X Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621616. [PMID: 39553953 PMCID: PMC11566027 DOI: 10.1101/2024.11.01.621616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited intellectual disability and often accompanied with debilitating pathologies including seizures and hyperactivity. FXS arises from a trinucleotide repeat expansion in the 5' UTR of the FMR1 gene that silences expression of the RNA-binding protein FMRP. Despite progress in understanding FMRP functions, the identification of effective therapeutic targets has lagged and at present there are no viable treatment options. Here we identify the α7 nicotinic acetylcholine receptor (nAChR) as candidate target for intervention in FXS. In the early postnatal hippocampus of Fmr1 knockout (KO) mice, an established pre-clinical model of FXS, the α7 nAChR accessory protein Ly6H is abnormally enriched at the neuronal surface and mislocalized in dendrites. Ly6H, a GPI-anchored protein, binds α7 nAChRs with high affinity and can limit α7 nAChR surface expression and signaling. We find that α7 nAChR-evoked Ca2+ responses are dampened in immature glutamatergic and GABAergic Fmr1 KO neurons compared to wild type. Knockdown of endogenous Ly6H in Fmr1 KO neurons is sufficient to rescue dampened α7 nAChR Ca2+ responses in vitro, providing evidence of a cell-autonomous role for Ly6H aberrant expression in α7 nAChR hypofunction. In line with intrinsic deficits in α7 nAChR activity in Fmr1 KO neurons, in vivo administration of the α7 nAChR-selective positive allosteric modulator PNU-120596 reduced hyperactivity and seizure severity in adolescent Fmr1 KO mice. Our mechanistic studies together with evidence of the in vivo efficacy of α7 nAChR augmentation implicate α7 nAChR hypofunction in FXS pathology.
Collapse
Affiliation(s)
- Sarah Goebel
- Department of Neuroscience, Albert Einstein College of Medicine; New York, NY, U.S.A
| | | | - Vytas K. Verselis
- Department of Neuroscience, Albert Einstein College of Medicine; New York, NY, U.S.A
| | - Anna Francesconi
- Department of Neuroscience, Albert Einstein College of Medicine; New York, NY, U.S.A
| |
Collapse
|
8
|
Guan Z. Alterations in Neuronal Nicotinic Acetylcholine Receptors in the Pathogenesis of Various Cognitive Impairments. CNS Neurosci Ther 2024; 30:e70069. [PMID: 39370620 PMCID: PMC11456617 DOI: 10.1111/cns.70069] [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: 03/16/2024] [Revised: 09/02/2024] [Accepted: 09/15/2024] [Indexed: 10/08/2024] Open
Abstract
Cognitive impairment is a typical symptom of both neurodegenerative and certain other diseases. In connection with these different pathologies, the etiology and neurological and metabolic changes associated with cognitive impairment must differ. Until these characteristics and differences are understood in greater detail, pharmacological treatment of the different forms of cognitive impairment remains suboptimal. Neurotransmitter receptors, including neuronal nicotinic acetylcholine receptors (nAChRs), dopamine receptors, and glutamine receptors, play key roles in the functions and metabolisms of the brain. Among these, the role of nAChRs in the development of cognitive impairment has attracted more and more attention. The present review summarizes what is presently known concerning the structure, distribution, metabolism, and function of nAChRs, as well as their involvement in major cognitive disorders such as Alzheimer's disease, Parkinson's disease, vascular dementia, schizophrenia, and diabetes mellitus. As will be discussed, the relevant scientific literature reveals clearly that the α4β2 and α7 nAChR subtypes and/or subunits of the receptors play major roles in maintaining cognitive function and in neuroprotection of the brain. Accordingly, focusing on these as targets of drug therapy can be expected to lead to breakthroughs in the treatment of cognitive disorders such as AD and schizophrenia.
Collapse
Affiliation(s)
- Zhi‐Zhong Guan
- Department of PathologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- Key Laboratory of Endemic and Ethnic DiseasesGuizhou Medical University, Ministry of Education and Provincial Key Laboratory of Medical Molecular BiologyGuiyangP.R. China
| |
Collapse
|
9
|
Brockmöller S, Molitor LM, Seeger T, Worek F, Rothmiller S. N-Glycosylation Deficiency in Transgene α7 nAChR and RIC3 Expressing CHO Cells Without NACHO. J Membr Biol 2024; 257:245-256. [PMID: 38967800 DOI: 10.1007/s00232-024-00317-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
The human neuronal nicotinic acetylcholine receptor α7 (nAChR) is an important target implicated in diseases like Alzheimer's or Parkinson's, as well as a validated target for drug discovery. For α7 nAChR model systems, correct folding and ion influx functions are essential. Two chaperones, resistance to inhibitors of cholinesterase 3 (RIC3) and novel nAChR regulator (NACHO), enhance the assembly and function of α7 nAChR. This study investigates the consequence of NACHO absence on α7 nAChR expression and function. Therefore, the sequences of human α7 nAChR and human RIC3 were transduced in Chinese hamster ovary (CHO) cells. Protein expression and function of α7 nAChR were confirmed by Western blot and voltage clamp, respectively. Cellular viability was assessed by cell proliferation and lactate dehydrogenase assays. Intracellular and extracellular expression were determined by in/on-cell Western, compared with another nAChR subtype by novel cluster fluorescence-linked immunosorbent assay, and N-glycosylation efficiency was assessed by glycosylation digest. The transgene CHO cell line showed expected protein expression and function for α7 nAChR and cell viability was barely influenced by overexpression. While intracellular levels of α7 nAChR were as anticipated, plasma membrane insertion was low. The glycosylation digest revealed no appreciable N-glycosylation product. This study demonstrates a stable and functional cell line expressing α7 nAChR, whose protein expression, function, and viability are not affected by the absence of NACHO. The reduced plasma membrane insertion of α7 nAChR, combined with incorrect matured N-glycosylation at the Golgi apparatus, suggests a loss of recognition signal for lectin sorting.
Collapse
Affiliation(s)
| | | | - Thomas Seeger
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| |
Collapse
|
10
|
Guo Z, Zhu AT, Wei X, Jiang Y, Yu Y, Noh I, Gao W, Fang RH, Zhang L. A genetically engineered neuronal membrane-based nanotoxoid elicits protective immunity against neurotoxins. Bioact Mater 2024; 38:321-330. [PMID: 38764446 PMCID: PMC11101676 DOI: 10.1016/j.bioactmat.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Given their dangerous effects on the nervous system, neurotoxins represent a significant threat to public health. Various therapeutic approaches, including chelating agents, receptor decoys, and toxin-neutralizing antibodies, have been explored. While prophylactic vaccines are desirable, it is oftentimes difficult to effectively balance their safety and efficacy given the highly dangerous nature of neurotoxins. To address this, we report here on a nanovaccine against neurotoxins that leverages the detoxifying properties of cell membrane-coated nanoparticles. A genetically modified cell line with constitutive overexpression of the α7 nicotinic acetylcholine receptor is developed as a membrane source to generate biomimetic nanoparticles that can effectively and irreversibly bind to α-bungarotoxin, a model neurotoxin. This abrogates the biological activity of the toxin, enabling the resulting nanotoxoid to be safely delivered into the body and processed by the immune system. When co-administered with an immunological adjuvant, a strong humoral response against α-bungarotoxin is generated that protects vaccinated mice against a lethal dose of the toxin. Overall, this work highlights the potential of using genetic modification strategies to develop nanotoxoid formulations against various biological threats.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Audrey T. Zhu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yiyan Yu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ilkoo Noh
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
11
|
Mazzaferro S, Kang G, Natarajan K, Hibbs RE, Sine SM. Structural bases for stoichiometry-selective calcium potentiation of a neuronal nicotinic receptor. Br J Pharmacol 2024; 181:1973-1992. [PMID: 38454578 PMCID: PMC12048133 DOI: 10.1111/bph.16321] [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: 09/13/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND AND PURPOSE α4β2 nicotinic acetylcholine (nACh) receptors assemble in two stoichiometric forms, one of which is potentiated by calcium. The sites of calcium binding that underpin potentiation are not known. EXPERIMENTAL APPROACH To identify calcium binding sites, we applied cryo-electron microscopy (cryo-EM) and molecular dynamics (MD) simulations to each stoichiometric form of the α4β2 nACh receptor in the presence of calcium ions. To test whether the identified calcium sites are linked to potentiation, we generated mutants of anionic residues at the sites, expressed wild type and mutant receptors in clonal mammalian fibroblasts, and recorded ACh-elicited single-channel currents with or without calcium. KEY RESULTS Both cryo-EM and MD simulations show calcium bound to a site between the extracellular and transmembrane domains of each α4 subunit (ECD-TMD site). Substituting alanine for anionic residues at the ECD-TMD site abolishes stoichiometry-selective calcium potentiation, as monitored by single-channel patch clamp electrophysiology. Additionally, MD simulation reveals calcium association at subunit interfaces within the extracellular domain. Substituting alanine for anionic residues at the ECD sites reduces or abolishes stoichiometry-selective calcium potentiation. CONCLUSIONS AND IMPLICATIONS Stoichiometry-selective calcium potentiation of the α4β2 nACh receptor is achieved by calcium association with topographically distinct sites framed by anionic residues within the α4 subunit and between the α4 and β2 subunits. Stoichiometry-selective calcium potentiation could result from the greater number of calcium sites in the stoichiometric form with three rather than two α4 subunits. The results are relevant to modulation of signalling via α4β2 nACh receptors in physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Simone Mazzaferro
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Guipeun Kang
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kathiresan Natarajan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan E Hibbs
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurobiology, University of California San Diego, La Jolla, California, USA
| | - Steven M Sine
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
- Department of Neurology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
12
|
Tsuzuki A, Yamasaki M, Konno K, Miyazaki T, Takei N, Tomita S, Yuzaki M, Watanabe M. Abundant extrasynaptic expression of α3β4-containing nicotinic acetylcholine receptors in the medial habenula-interpeduncular nucleus pathway in mice. Sci Rep 2024; 14:14193. [PMID: 38902419 PMCID: PMC11189931 DOI: 10.1038/s41598-024-65076-3] [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: 03/15/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) in the medial habenula (MHb)-interpeduncular nucleus (IPN) pathway play critical roles in nicotine-related behaviors. This pathway is particularly enriched in nAChR α3 and β4 subunits, both of which are genetically linked to nicotine dependence. However, the cellular and subcellular expression of endogenous α3β4-containing nAChRs remains largely unknown because specific antibodies and appropriate detection methods were unavailable. Here, we successfully uncovered the expression of endogenous nAChRs containing α3 and β4 subunits in the MHb-IPN pathway using novel specific antibodies and a fixative glyoxal that enables simultaneous detection of synaptic and extrasynaptic molecules. Immunofluorescence and immunoelectron microscopy revealed that both subunits were predominantly localized to the extrasynaptic cell surface of somatodendritic and axonal compartments of MHb neurons but not at their synaptic junctions. Immunolabeling for α3 and β4 subunits disappeared in α5β4-knockout brains, which we used as negative controls. The enriched and diffuse extrasynaptic expression along the MHb-IPN pathway suggests that α3β4-containing nAChRs may enhance the excitability of MHb neurons and neurotransmitter release from their presynaptic terminals in the IPN. The revealed distribution pattern provides a molecular and anatomical basis for understanding the functional role of α3β4-containing nAChRs in the crucial pathway of nicotine dependence.
Collapse
Grants
- 17KK0160 Ministry of Education, Culture, Sports, Science and Technology
- 21K06746 Ministry of Education, Culture, Sports, Science and Technology
- 22K06784 Ministry of Education, Culture, Sports, Science and Technology
- 20H05628 Ministry of Education, Culture, Sports, Science and Technology
- 20H05628 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Asuka Tsuzuki
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan.
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Taisuke Miyazaki
- Department of Functioning and Disability, Faculty of Health Sciences, Hokkaido University, Sapporo, 060-8638, Japan
| | - Norio Takei
- Institute for Animal Experimentation, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Department of Neuroscience, and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Michisuke Yuzaki
- Department of Physiology, School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| |
Collapse
|
13
|
Gotti C, Clementi F, Zoli M. Auxiliary protein and chaperone regulation of neuronal nicotinic receptor subtype expression and function. Pharmacol Res 2024; 200:107067. [PMID: 38218358 DOI: 10.1016/j.phrs.2024.107067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Neuronal nicotinic acetylcholine receptors (nAChRs) are a family of pentameric, ligand-gated ion channels that are located on the surface of neurons and non-neuronal cells and have multiple physiological and pathophysiological functions. In order to reach the cell surface, many nAChR subtypes require the help of chaperone and/or auxiliary/accessory proteins for their assembly, trafficking, pharmacological modulation, and normal functioning in vivo. The use of powerful genome-wide cDNA screening has led to the identification and characterisation of the molecules and mechanisms that participate in the assembly and trafficking of receptor subtypes, including chaperone and auxiliary or accessory proteins. The aim of this review is to describe the latest findings concerning nAChR chaperones and auxiliary proteins and pharmacological chaperones, and how some of them control receptor biogenesis or regulate channel activation and pharmacology. Some auxiliary proteins are subtype selective, some regulate various subtypes, and some not only modulate nAChRs but also target other receptors and signalling pathways. We also discuss how changes in auxiliary proteins may be involved in nAChR dysfunctions.
Collapse
Affiliation(s)
- Cecilia Gotti
- CNR, Institute of Neuroscience, Milan, Italy; NeuroMi Milan Center for Neuroscience, University of Milano-Bicocca, Italy.
| | - Francesco Clementi
- CNR, Institute of Neuroscience, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology (CfNN), University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Sansone L, Milani F, Fabrizi R, Belli M, Cristina M, Zagà V, de Iure A, Cicconi L, Bonassi S, Russo P. Nicotine: From Discovery to Biological Effects. Int J Mol Sci 2023; 24:14570. [PMID: 37834017 PMCID: PMC10572882 DOI: 10.3390/ijms241914570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Nicotine, the primary psychoactive agent in tobacco leaves, has led to the widespread use of tobacco, with over one billion smokers globally. This article provides a historical overview of tobacco and discusses tobacco dependence, as well as the biological effects induced by nicotine on mammalian cells. Nicotine induces various biological effects, such as neoangiogenesis, cell division, and proliferation, and it affects neural and non-neural cells through specific pathways downstream of nicotinic receptors (nAChRs). Specific effects mediated by α7 nAChRs are highlighted. Nicotine is highly addictive and hazardous. Public health initiatives should prioritize combating smoking and its associated risks. Understanding nicotine's complex biological effects is essential for comprehensive research and informed health policies. While potential links between nicotine and COVID-19 severity warrant further investigation, smoking remains a significant cause of morbidity and mortality globally. Effective public health strategies are vital to promote healthier lifestyles.
Collapse
Affiliation(s)
- Luigi Sansone
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| | - Francesca Milani
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Riccardo Fabrizi
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Manuel Belli
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| | - Mario Cristina
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Department of Molecular Medicine, University La Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Zagà
- Italian Society of Tabaccology (SITAB), 00136 Bologna, Italy;
| | - Antonio de Iure
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Experimental Neurophysiology IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Luca Cicconi
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Stefano Bonassi
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Patrizia Russo
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| |
Collapse
|
15
|
Burns LH, Pei Z, Wang HY. Targeting α7 nicotinic acetylcholine receptors and their protein interactions in Alzheimer's disease drug development. Drug Dev Res 2023; 84:1085-1095. [PMID: 37291958 DOI: 10.1002/ddr.22085] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/09/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
The decades-old cholinergic hypothesis of Alzheimer's disease (AD) led to clinical testing and FDA approval of acetylcholinesterase inhibitor drugs. Subsequently, the α7 nicotinic acetylcholine receptor (α7nAChR) was proposed as a new drug target for enhancing cholinergic neurotransmission. Nearly simultaneously, soluble amyloid β1-42 (Aβ42 ) was shown to bind α7nAChR with picomolar affinity to activate kinases that hyperphosphorylate tau, the precursor to tau-containing tangles. Multiple biopharmaceutical companies explored α7nAChR as a drug target for AD, mostly to enhance neurotransmission. Directly targeting α7nAChR proved to be a drug development challenge. The ultra-high-affinity interaction between Aβ42 and α7nAChR posed a significant hurdle for direct competition in the AD brain. The receptor rapidly desensitizes, undermining efficacy of agonists. Drug discovery approaches therefore included partial agonists and allosteric modulators of α7nAChR. After substantial effort, numerous drug candidates were abandoned due to lack of efficacy or drug-related toxicities. As alternatives, proteins interacting with α7nAChR were sought. In 2016, a novel nAChR regulator was identified, but no drug candidates have emerged from this effort. In 2012, the interaction of filamin A with α7nAChR was shown to be critical to Aβ42 's toxic signaling via α7nAChR, presenting a new drug target. The novel drug candidate simufilam disrupts the filamin A-α7nAChR interaction, reduces Aβ42 's high-affinity binding to α7nAChR, and suppresses Aβ42 's toxic signaling. Early clinical trials of simufilam showed improvements in experimental CSF biomarkers and indications of cognitive improvement in mild AD patients at 1 year. Simufilam is currently in phase 3 clinical trials as a disease-modifying treatment for AD.
Collapse
Affiliation(s)
| | - Zhe Pei
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, New York, USA
| | - Hoau-Yan Wang
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, New York, USA
- Department of Biology and Neuroscience, Graduate School of the City University of New York, New York, New York, USA
| |
Collapse
|
16
|
Rocha JJ, Jayaram SA, Stevens TJ, Muschalik N, Shah RD, Emran S, Robles C, Freeman M, Munro S. Functional unknomics: Systematic screening of conserved genes of unknown function. PLoS Biol 2023; 21:e3002222. [PMID: 37552676 PMCID: PMC10409296 DOI: 10.1371/journal.pbio.3002222] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/27/2023] [Indexed: 08/10/2023] Open
Abstract
The human genome encodes approximately 20,000 proteins, many still uncharacterised. It has become clear that scientific research tends to focus on well-studied proteins, leading to a concern that poorly understood genes are unjustifiably neglected. To address this, we have developed a publicly available and customisable "Unknome database" that ranks proteins based on how little is known about them. We applied RNA interference (RNAi) in Drosophila to 260 unknown genes that are conserved between flies and humans. Knockdown of some genes resulted in loss of viability, and functional screening of the rest revealed hits for fertility, development, locomotion, protein quality control, and resilience to stress. CRISPR/Cas9 gene disruption validated a component of Notch signalling and 2 genes contributing to male fertility. Our work illustrates the importance of poorly understood genes, provides a resource to accelerate future research, and highlights a need to support database curation to ensure that misannotation does not erode our awareness of our own ignorance.
Collapse
Affiliation(s)
- João J. Rocha
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | - Tim J. Stevens
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | - Rajen D. Shah
- Centre for Mathematical Sciences, University of Cambridge, Cambridge, United Kingdom
| | - Sahar Emran
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Cristina Robles
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Matthew Freeman
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
17
|
Khalfaoui L, Mukhtasimova N, Kelley B, Wells N, Teske JJ, Roos BB, Borkar NA, Zhang EY, Sine SM, Prakash YS, Pabelick CM. Functional α7 nicotinic receptors in human airway smooth muscle increase intracellular calcium concentration and contractility in asthmatics. Am J Physiol Lung Cell Mol Physiol 2023; 325:L17-L29. [PMID: 37192375 PMCID: PMC10292984 DOI: 10.1152/ajplung.00260.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 05/18/2023] Open
Abstract
Although nicotinic acetylcholine receptors (nAChRs) are commonly associated with neurons in the brain and periphery, recent data indicate that they are also expressed in non-neuronal tissues. We recently found the alpha7 (α7nAChR) subunit is highly expressed in human airway smooth muscle (hASM) with substantial increase in asthmatics, but their functionality remains unknown. We investigated the location and functional role of α7nAChRs in hASM cells from normal versus mild-moderate asthmatic patients. Immunostaining and protein analyses showed α7nAChR in the plasma membrane including in asthmatics. In asthmatic hASM, patch-clamp recordings revealed significantly higher functional homomeric α7nAChR channels. Real-time fluorescence imaging showed nicotine, via α7nAChR, increases intracellular Ca2+ ([Ca2+]i) independent of ACh effects, particularly in asthmatic hASM, while cellular traction force microscopy showed nicotine-induced contractility including in asthmatics. These results indicate functional homomeric and heteromeric nAChRs that are increased in asthmatic hASM, with pharmacology that likely differ owing to different subunit interfaces that form the orthosteric sites. nAChRs may represent a novel target in alleviating airway hyperresponsiveness in asthma.NEW & NOTEWORTHY Cigarette smoking and vaping exacerbate asthma. Understanding the mechanisms of nicotine effects in asthmatic airways is important. This study demonstrates that functional alpha7 nicotinic acetylcholine receptors (α7nAChRs) are expressed in human airway smooth muscle, including from asthmatics, and enhance intracellular calcium and contractility. Although a7nAChRs are associated with neuronal pathways, α7nAChR in smooth muscle suggests inhaled nicotine (e.g., vaping) can directly influence airway contractility. Targeting α7nAChR may represent a novel approach to alleviating airway hyperresponsiveness in asthma.
Collapse
Affiliation(s)
- Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Nuriya Mukhtasimova
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Brian Kelley
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Natalya Wells
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Jacob J Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Niyati A Borkar
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Emily Y Zhang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Steven M Sine
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
18
|
York JM, Borghese CM, George AA, Cannatella DC, Zakon HH. A potential cost of evolving epibatidine resistance in poison frogs. BMC Biol 2023; 21:144. [PMID: 37370119 DOI: 10.1186/s12915-023-01637-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Some dendrobatid poison frogs sequester the toxin epibatidine as a defense against predators. We previously identified an amino acid substitution (S108C) at a highly conserved site in a nicotinic acetylcholine receptor β2 subunit of dendrobatid frogs that decreases sensitivity to epibatidine in the brain-expressing α4β2 receptor. Introduction of S108C to the orthologous high-sensitivity human receptor similarly decreased sensitivity to epibatidine but also decreased sensitivity to acetylcholine, a potential cost if this were to occur in dendrobatids. This decrease in the acetylcholine sensitivity manifested as a biphasic acetylcholine concentration-response curve consistent with the addition of low-sensitivity receptors. Surprisingly, the addition of the β2 S108C into the α4β2 receptor of the dendrobatid Epipedobates anthonyi did not change acetylcholine sensitivity, appearing cost-free. We proposed that toxin-bearing dendrobatids may have additional amino acid substitutions protecting their receptors from alterations in acetylcholine sensitivity. To test this, in the current study, we compared the dendrobatid receptor to its homologs from two non-dendrobatid frogs. RESULTS The introduction of S108C into the α4β2 receptors of two non-dendrobatid frogs also does not affect acetylcholine sensitivity suggesting no additional dendrobatid-specific substitutions. However, S108C decreased the magnitude of neurotransmitter-induced currents in Epipedobates and the non-dendrobatid frogs. We confirmed that decreased current resulted from fewer receptors in the plasma membrane in Epipedobates using radiolabeled antibodies against the receptors. To test whether S108C alteration of acetylcholine sensitivity in the human receptor was due to (1) adding low-sensitivity binding sites by changing stoichiometry or (2) converting existing high- to low-sensitivity binding sites with no stoichiometric alteration, we made concatenated α4β2 receptors in stoichiometry with only high-sensitivity sites. S108C substitutions decreased maximal current and number of immunolabeled receptors but no longer altered acetylcholine sensitivity. CONCLUSIONS The most parsimonious explanation of our current and previous work is that the S108C substitution renders the β2 subunit less efficient in assembling/trafficking, thereby decreasing the number of receptors in the plasma membrane. Thus, while β2 S108C protects dendrobatids against sequestered epibatidine, it incurs a potential physiological cost of disrupted α4β2 receptor function.
Collapse
Affiliation(s)
- Julia M York
- Department of Neuroscience, The University of Texas, Austin, TX, USA
- Department of Integrative Biology, and Biodiversity Center, The University of Texas, Austin, TX, USA
| | | | - Andrew A George
- Department of Neurobiology, The Barrow Neurological Institute, Phoenix, AZ, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - David C Cannatella
- Department of Integrative Biology, and Biodiversity Center, The University of Texas, Austin, TX, USA
| | - Harold H Zakon
- Department of Neuroscience, The University of Texas, Austin, TX, USA.
- Department of Integrative Biology, and Biodiversity Center, The University of Texas, Austin, TX, USA.
| |
Collapse
|
19
|
Maldifassi MC, Rego Campello H, Gallagher T, Lester HA, Dougherty DA. Human α6 β4 Nicotinic Acetylcholine Receptor: Heterologous Expression and Agonist Behavior Provide Insights into the Immediate Binding Site. Mol Pharmacol 2023; 103:339-347. [PMID: 37001996 DOI: 10.1124/molpharm.123.000672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Study of α6β4 nicotinic acetylcholine receptors (nAChRs) as a pharmacological target has recently gained interest because of their involvement in analgesia, control of catecholamine secretion, dopaminergic pathways, and aversive pathways. However, an extensive characterization of the human α6β4 nAChRs has been vitiated by technical difficulties resulting in poor receptor expression. In 2020, Knowland and collaborators identified BARP (β-anchoring and regulatory protein), a previously known voltage-gated calcium channel suppressor, as a novel human α6β4 chaperone. Here, we establish that co-expression of human BARP with human α6β4 in Xenopus oocytes, resulted in the functional expression of human α6β4 receptors with acetylcholine-elicited currents that allow an in-depth characterization of the receptor using two electrode voltage-clamp electrophysiology together with diverse agonists and receptor mutations. We report: 1) an extended pharmacological characterization of the receptor, and 2) key residues for agonist-activity located in or near the first shell of the binding pocket. SIGNIFICANCE STATEMENT: The human α6β4 nicotinic acetylcholine receptor has attained increased interest because of its involvement in diverse physiological processes and diseases. Although recognized as a pharmacological target, development of specific agonists has been hampered by limited knowledge of its structural characteristics and by challenges in expressing the receptor. By including the chaperone β-anchoring and regulatory protein for enhanced expression and employing different ligands, we have studied the pharmacology of α6β4, providing insight into receptor residues and structural requirements for ligands important to consider for agonist-induced activation.
Collapse
Affiliation(s)
- María Constanza Maldifassi
- Divisions of Chemistry and Chemical Engineering (M.C.M., D.A.D.) and Biology and Biological Engineering (H.A.L.), California Institute of Technology, Pasadena, California; and School of Chemistry, University of Bristol, Bristol, United Kingdom (H.R.C., T.G.)
| | - Hugo Rego Campello
- Divisions of Chemistry and Chemical Engineering (M.C.M., D.A.D.) and Biology and Biological Engineering (H.A.L.), California Institute of Technology, Pasadena, California; and School of Chemistry, University of Bristol, Bristol, United Kingdom (H.R.C., T.G.)
| | - Timothy Gallagher
- Divisions of Chemistry and Chemical Engineering (M.C.M., D.A.D.) and Biology and Biological Engineering (H.A.L.), California Institute of Technology, Pasadena, California; and School of Chemistry, University of Bristol, Bristol, United Kingdom (H.R.C., T.G.)
| | - Henry A Lester
- Divisions of Chemistry and Chemical Engineering (M.C.M., D.A.D.) and Biology and Biological Engineering (H.A.L.), California Institute of Technology, Pasadena, California; and School of Chemistry, University of Bristol, Bristol, United Kingdom (H.R.C., T.G.)
| | - Dennis A Dougherty
- Divisions of Chemistry and Chemical Engineering (M.C.M., D.A.D.) and Biology and Biological Engineering (H.A.L.), California Institute of Technology, Pasadena, California; and School of Chemistry, University of Bristol, Bristol, United Kingdom (H.R.C., T.G.)
| |
Collapse
|
20
|
Pechlivanidou M, Ninou E, Karagiorgou K, Tsantila A, Mantegazza R, Francesca A, Furlan R, Dudeck L, Steiner J, Tzartos J, Tzartos S. Autoimmunity to Neuronal Nicotinic Acetylcholine Receptors. Pharmacol Res 2023; 192:106790. [PMID: 37164280 DOI: 10.1016/j.phrs.2023.106790] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are widely expressed in many and diverse cell types, participating in various functions of cells, tissues and systems. In this review, we focus on the autoimmunity against neuronal nAChRs, the specific autoantibodies and their mechanisms of pathological action in selected autoimmune diseases. We summarize the current relevant knowledge from human diseases as well as from experimental models of autoimmune neurological disorders related to antibodies against neuronal nAChR subunits. Despite the well-studied high immunogenicity of the muscle nAChRs where autoantibodies are the main pathogen of myasthenia gravis, autoimmunity to neuronal nAChRs seems infrequent, except for the autoantibodies to the ganglionic receptor, the α3 subunit containing nAChR (α3-nAChR), which are detected and are likely pathogenic in Autoimmune Autonomic Ganglionopathy (AAG). We describe the detection, presence and function of these antibodies and especially the recent development of a cell-based assay (CBA) which, contrary to until recently available assays, is highly specific for AAG. Rare reports of autoantibodies to the other neuronal nAChR subtypes include a few cases of antibodies to α7 and/or α4β2 nAChRs in Rasmussen encephalitis, schizophrenia, autoimmune meningoencephalomyelitis, and in some myasthenia gravis patients with concurrent CNS symptoms. Neuronal-type nAChRs are also present in several non-excitable tissues, however the presence and possible role of antibodies against them needs further verification. It is likely that the future development of more sensitive and disease-specific assays would reveal that neuronal nAChR autoantibodies are much more frequent and may explain the mechanisms of some seronegative autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Katerina Karagiorgou
- Tzartos NeuroDiagnostics, Athens, Greece; Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | | | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Andreetta Francesca
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaello Furlan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Rozzano, Milan, Italy; Clinical and Research Center - IRCCS, Humanitas University, Rozzano, Milan, Italy
| | - Leon Dudeck
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Health and Medical Prevention (CHaMP), Magdeburg, Germany; German Center for Mental Health DZPG, Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health C-I-R-C, Halle-Jena-Magdeburg, Germany
| | - John Tzartos
- 2(nd) Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece.
| | - Socrates Tzartos
- Tzartos NeuroDiagnostics, Athens, Greece; Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece; Department of Pharmacy, University of Patras, Patras, Greece.
| |
Collapse
|
21
|
Becchetti A, Grandi LC, Cerina M, Amadeo A. Nicotinic acetylcholine receptors and epilepsy. Pharmacol Res 2023; 189:106698. [PMID: 36796465 DOI: 10.1016/j.phrs.2023.106698] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Despite recent advances in understanding the causes of epilepsy, especially the genetic, comprehending the biological mechanisms that lead to the epileptic phenotype remains difficult. A paradigmatic case is constituted by the epilepsies caused by altered neuronal nicotinic acetylcholine receptors (nAChRs), which exert complex physiological functions in mature as well as developing brain. The ascending cholinergic projections exert potent control of forebrain excitability, and wide evidence implicates nAChR dysregulation as both cause and effect of epileptiform activity. First, tonic-clonic seizures are triggered by administration of high doses of nicotinic agonists, whereas non-convulsive doses have kindling effects. Second, sleep-related epilepsy can be caused by mutations on genes encoding nAChR subunits widely expressed in the forebrain (CHRNA4, CHRNB2, CHRNA2). Third, in animal models of acquired epilepsy, complex time-dependent alterations in cholinergic innervation are observed following repeated seizures. Heteromeric nAChRs are central players in epileptogenesis. Evidence is wide for autosomal dominant sleep-related hypermotor epilepsy (ADSHE). Studies of ADSHE-linked nAChR subunits in expression systems suggest that the epileptogenic process is promoted by overactive receptors. Investigation in animal models of ADSHE indicates that expression of mutant nAChRs can lead to lifelong hyperexcitability by altering i) the function of GABAergic populations in the mature neocortex and thalamus, ii) synaptic architecture during synaptogenesis. Understanding the balance of the epileptogenic effects in adult and developing networks is essential to plan rational therapy at different ages. Combining this knowledge with a deeper understanding of the functional and pharmacological properties of individual mutations will advance precision and personalized medicine in nAChR-dependent epilepsy.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Laura Clara Grandi
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Marta Cerina
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Alida Amadeo
- Department of Biosciences, University of Milano, Via Celoria 26, Milano 20133, Italy.
| |
Collapse
|
22
|
York JM, Borghese CM, George AA, Cannatella DC, Zakon HH. A potential cost of evolving epibatidine resistance in poison frogs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522789. [PMID: 36711899 PMCID: PMC9882002 DOI: 10.1101/2023.01.04.522789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Some poison arrow frogs sequester the toxin epibatidine as a defense against predators. We previously identified a single amino acid substitution (S108C) at a highly conserved site in a neuronal nicotinic acetylcholine receptor (nAChR) ß2 subunit that prevents epibatidine from binding to this receptor. When placed in a homologous mammalian nAChR this substitution minimized epibatidine binding but also perturbed acetylcholine binding, a clear cost. However, in the nAChRs of poison arrow frogs, this substitution appeared to have no detrimental effect on acetylcholine binding and, thus, appeared cost-free. Results The introduction of S108C into the α4β2 nAChRs of non-dendrobatid frogs also does not affect ACh sensitivity, when these receptors are expressed in Xenopus laevis oocytes. However, α4β2 nAChRs with C108 had a decreased magnitude of neurotransmitter-induced currents in all species tested ( Epipedobates anthonyi , non-dendrobatid frogs, as well as human), compared with α4β2 nAChRs with the conserved S108. Immunolabeling of frog or human α4β2 nAChRs in the plasma membrane using radiolabeled antibody against the β2 nAChR subunit shows that C108 significantly decreased the number of cell-surface α4β2 nAChRs, compared with S108. Conclusions While S108C protects these species against sequestered epibatidine, it incurs a potential physiological cost of disrupted α4β2 nAChR function. These results may explain the high conservation of a serine at this site in vertebrates, as well as provide an example of a tradeoff between beneficial and deleterious effects of an evolutionary change. They also provide important clues for future work on assembly and trafficking of this important neurotransmitter receptor.
Collapse
|
23
|
Speculation on How RIC-3 and Other Chaperones Facilitate α7 Nicotinic Receptor Folding and Assembly. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144527. [PMID: 35889400 PMCID: PMC9318448 DOI: 10.3390/molecules27144527] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022]
Abstract
The process of how multimeric transmembrane proteins fold and assemble in the endoplasmic reticulum is not well understood. The alpha7 nicotinic receptor (α7 nAChR) is a good model for multimeric protein assembly since it has at least two independent and specialized chaperones: Resistance to Inhibitors of Cholinesterase 3 (RIC-3) and Nicotinic Acetylcholine Receptor Regulator (NACHO). Recent cryo-EM and NMR data revealed structural features of α7 nAChRs. A ser-ala-pro (SAP) motif precedes a structurally important but unique "latch" helix in α7 nAChRs. A sampling of α7 sequences suggests the SAP motif is conserved from C. elegans to humans, but the latch sequence is only conserved in vertebrates. How RIC-3 and NACHO facilitate receptor subunits folding into their final pentameric configuration is not known. The artificial intelligence program AlphaFold2 recently predicted structures for NACHO and RIC-3. NACHO is highly conserved in sequence and structure across species, but RIC-3 is not. This review ponders how different intrinsically disordered RIC-3 isoforms from C. elegans to humans interact with α7 nAChR subunits despite having little sequence homology across RIC-3 species. Two models from the literature about how RIC-3 assists α7 nAChR assembly are evaluated considering recent structural information about the receptor and its chaperones.
Collapse
|
24
|
Belanger-Coast MG, Zhang M, Bugay V, Gutierrez RA, Gregory SR, Yu W, Brenner R. Dequalinium chloride is an antagonists of α7 nicotinic acetylcholine receptors. Eur J Pharmacol 2022; 925:175000. [PMID: 35525312 DOI: 10.1016/j.ejphar.2022.175000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022]
Abstract
Dequalinium chloride has been used primarily as antiseptic compounds, but recently has been investigated for its effects on specific targets, including muscarinic acetylcholine receptors. Here we investigated dequalinium chloride as an antagonist to α7 nicotinic acetylcholine receptors. The pharmacological properties of dequalinium were established using cell lines stably co-transfected with the calcium-permeable human α7 nicotinic acetylcholine receptors and its chaperone NACHO, calcium dye fluorescent measurements or a calcium-sensitive protein reporter, and patch clamp recording of ionic currents. Using calcium dye fluorescence plate reader measurements, we find dequalinium chloride is an antagonist of α7 nicotinic acetylcholine receptors with an IC50 of 672 nM in response to activation with 500 μM acetylcholine chloride and positive allosteric modulator PNU-120596. However, using a membrane-tethered GCAMP7s calcium reporter allowed detection of α7-mediated calcium flux in the absence of PNU-120596. Using this approach revealed an IC50 of 157 nM for dequalinium on 300 μM acetylcholine-evoked currents. Using patch clamp recordings with 300 μM acetylcholine chloride and 10 μM PNU-120596, we find lower concentrations are sufficient to block ionic currents, with IC50 of 120 nM for dequalinium chloride and 54 nM for the related UCL 1684 compound. In summary, we find that dequalinium chloride and UCL1684, which are generally used to block SK-type potassium channels, are also highly effective antagonists of α7 nicotinic acetylcholine receptors. This finding, in combination with previous studies of muscarinic acetylcholine receptors, clearly establishes dequalinium compounds within the class of general anti-cholinergic antagonists.
Collapse
Affiliation(s)
- Matthieu G Belanger-Coast
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mei Zhang
- Sophion Bioscience, Inc, 400 Trade Center Drive, Suite, 6900, Woburn, MA, USA
| | - Vladislav Bugay
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Raul A Gutierrez
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Summer R Gregory
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Weifeng Yu
- Sophion Bioscience, Inc, 400 Trade Center Drive, Suite, 6900, Woburn, MA, USA
| | - Robert Brenner
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
25
|
Karagiorgou K, Dandoulaki M, Mantegazza R, Andreetta F, Furlan R, Lindstrom J, Zisimopoulou P, Chroni E, Kokotis P, Anagnostou E, Tzanetakos D, Breza M, Katsarou Z, Amoiridis G, Mastorodemos V, Bregianni M, Bonakis A, Tsivgoulis G, Voumvourakis K, Tzartos S, Tzartos J. Novel Cell-Based Assay for Alpha-3 Nicotinic Receptor Antibodies Detects Antibodies Exclusively in Autoimmune Autonomic Ganglionopathy. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/3/e1162. [PMID: 35351814 PMCID: PMC8969289 DOI: 10.1212/nxi.0000000000001162] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022]
Abstract
Background and Objectives Autoantibodies against α3-subunit–containing nicotinic acetylcholine receptors (α3-nAChRs), usually measured by radioimmunoprecipitation assay (RIPA), are detected in patients with autoimmune autonomic ganglionopathy (AAG). However, low α3-nAChR antibody levels are frequently detected in other neurologic diseases with questionable significance. Our objective was to develop a method for the selective detection of the potentially pathogenic α3-nAChR antibodies, seemingly present only in patients with AAG. Methods The study involved sera from 55 patients from Greece, suspected for autonomic failure, and 13 patients from Italy diagnosed with autonomic failure, positive for α3-nAChR antibodies by RIPA. In addition, sera from 52 patients with Ca2+ channel or Hu antibodies and from 2,628 controls with various neuroimmune diseases were included. A sensitive live cell-based assay (CBA) with α3-nAChR–transfected cells was developed to detect antibodies against the cell-exposed α3-nAChR domain. Results Twenty-five patients were found α3-nAChR antibody positive by RIPA. Fifteen of 25 patients were also CBA positive. Of interest, all 15 CBA-positive patients had AAG, whereas all 10 CBA-negative patients had other neurologic diseases. RIPA antibody levels of the CBA-negative sera were low, although our CBA could detect dilutions of AAG sera corresponding to equally low RIPA antibody levels. No serum bound to control-transfected cells, and none of the 2,628 controls was α3-CBA positive. Discussion This study showed that in contrast to the established RIPA for α3-nAChR antibodies, which at low levels is of moderate disease specificity, our CBA seems AAG specific, while at least equally sensitive with the RIPA. This study provides Class II evidence that α3-nAChR CBA is a specific assay for AAG. Classification of Evidence This study provides Class II evidence that an α3-nAChR cell-based assay is a more specific assay for AAG than the standard RIPA.
Collapse
Affiliation(s)
- Katerina Karagiorgou
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Maria Dandoulaki
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Renato Mantegazza
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Francesca Andreetta
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Raffaello Furlan
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Jon Lindstrom
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Paraskevi Zisimopoulou
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Elisabeth Chroni
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Panagiotis Kokotis
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Evangelos Anagnostou
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Dimitrios Tzanetakos
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Marianthi Breza
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Zoe Katsarou
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Georgios Amoiridis
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Vasileios Mastorodemos
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Marianna Bregianni
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Anastasios Bonakis
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Georgios Tsivgoulis
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Konstantinos Voumvourakis
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - Socrates Tzartos
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| | - John Tzartos
- From the Tzartos NeuroDiagnostics (K.K., M.D., S.T., J.T.), Athens; Department of Biochemistry and Biotechnology (K.K.), University of Thessaly, Larissa, Greece; Neuroimmunology and Neuromuscular Diseases Unit (R.M., F.A.), Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy; Department of Biomedical Sciences Humanitas University (R.F.), Milan, Italy; Department of Neuroscience (J.L.), Medical School, University of Pennsylvania, Philadelphia, PA; Department of Neurobiology (P.Z., S.T.), Hellenic Pasteur Institute, Athens, Greece; Department of Neurology (E.C.), School of Medicine, University of Patras; 1st Department of Neurology (P.K., E.A., D.T., M. Breza), School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens (NKUA), Athens; Department of Neurology (Z.K.), Hippokration Hospital, Thessaloniki; Neurology Department (G.A., V.M.), University Hospital of Crete, Heraklion, Crete; and 2nd Department of Neurology (M. Bregianni, A.B., G.T., K.V., J.T.), Attikon University Hospital, School of Medicine, NKUA, Athens, Greece
| |
Collapse
|
26
|
Nicotinic Acetylcholine Receptors and Microglia as Therapeutic and Imaging Targets in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092780. [PMID: 35566132 PMCID: PMC9102429 DOI: 10.3390/molecules27092780] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) accumulation and tauopathy are considered the pathological hallmarks of Alzheimer’s disease (AD), but attenuation in choline signaling, including decreased nicotinic acetylcholine receptors (nAChRs), is evident in the early phase of AD. Currently, there are no drugs that can suppress the progression of AD due to a limited understanding of AD pathophysiology. For this, diagnostic methods that can assess disease progression non-invasively before the onset of AD symptoms are essential, and it would be valuable to incorporate the concept of neurotheranostics, which simultaneously enables diagnosis and treatment. The neuroprotective pathways activated by nAChRs are attractive targets as these receptors may regulate microglial-mediated neuroinflammation. Microglia exhibit both pro- and anti-inflammatory functions that could be modulated to mitigate AD pathogenesis. Currently, single-cell analysis is identifying microglial subpopulations that may have specific functions in different stages of AD pathologies. Thus, the ability to image nAChRs and microglia in AD according to the stage of the disease in the living brain may lead to the development of new diagnostic and therapeutic methods. In this review, we summarize and discuss the recent findings on the nAChRs and microglia, as well as their methods for live imaging in the context of diagnosis, prophylaxis, and therapy for AD.
Collapse
|
27
|
Lipovsek M, Marcovich I, Elgoyhen AB. The Hair Cell α9α10 Nicotinic Acetylcholine Receptor: Odd Cousin in an Old Family. Front Cell Neurosci 2021; 15:785265. [PMID: 34867208 PMCID: PMC8634148 DOI: 10.3389/fncel.2021.785265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are a subfamily of pentameric ligand-gated ion channels with members identified in most eumetazoan clades. In vertebrates, they are divided into three subgroups, according to their main tissue of expression: neuronal, muscle and hair cell nAChRs. Each receptor subtype is composed of different subunits, encoded by paralogous genes. The latest to be identified are the α9 and α10 subunits, expressed in the mechanosensory hair cells of the inner ear and the lateral line, where they mediate efferent modulation. α9α10 nAChRs are the most divergent amongst all nicotinic receptors, showing marked differences in their degree of sequence conservation, their expression pattern, their subunit co-assembly rules and, most importantly, their functional properties. Here, we review recent advances in the understanding of the structure and evolution of nAChRs. We discuss the functional consequences of sequence divergence and conservation, with special emphasis on the hair cell α9α10 receptor, a seemingly distant cousin of neuronal and muscle nicotinic receptors. Finally, we highlight potential links between the evolution of the octavolateral system and the extreme divergence of vertebrate α9α10 receptors.
Collapse
Affiliation(s)
- Marcela Lipovsek
- Ear Institute, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Irina Marcovich
- Departments of Otolaryngology & Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
28
|
Batool S, Akhter B, Zaidi J, Visser F, Petrie G, Hill M, Syed NI. Neuronal Menin Overexpression Rescues Learning and Memory Phenotype in CA1-Specific α7 nAChRs KD Mice. Cells 2021; 10:3286. [PMID: 34943798 PMCID: PMC8699470 DOI: 10.3390/cells10123286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 01/08/2023] Open
Abstract
The perturbation of nicotinic cholinergic receptors is thought to underlie many neurodegenerative and neuropsychiatric disorders, such as Alzheimer's and schizophrenia. We previously identified that the tumor suppressor gene, MEN1, regulates both the expression and synaptic targeting of α7 nAChRs in the mouse hippocampal neurons in vitro. Here we sought to determine whether the α7 nAChRs gene expression reciprocally regulates the expression of menin, the protein encoded by the MEN1 gene, and if this interplay impacts learning and memory. We demonstrate here that α7 nAChRs knockdown (KD) both in in vitro and in vivo, initially upregulated and then subsequently downregulated menin expression. Exogenous expression of menin using an AAV transduction approach rescued α7 nAChRs KD mediated functional and behavioral deficits specifically in hippocampal (CA1) neurons. These effects involved the modulation of the α7 nAChR subunit expression and functional clustering at the synaptic sites. Our data thus demonstrates a novel and important interplay between the MEN1 gene and the α7 nAChRs in regulating hippocampal-dependent learning and memory.
Collapse
Affiliation(s)
- Shadab Batool
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.B.); (B.A.); (F.V.); (G.P.); (M.H.)
- Department of Neuroscience, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Basma Akhter
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.B.); (B.A.); (F.V.); (G.P.); (M.H.)
| | - Jawwad Zaidi
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Frank Visser
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.B.); (B.A.); (F.V.); (G.P.); (M.H.)
| | - Gavin Petrie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.B.); (B.A.); (F.V.); (G.P.); (M.H.)
- Department of Neuroscience, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Matthew Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.B.); (B.A.); (F.V.); (G.P.); (M.H.)
| | - Naweed I. Syed
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
29
|
Wu M, Liu CZ, Barrall EA, Rissman RA, Joiner WJ. Unbalanced Regulation of α7 nAChRs by Ly6h and NACHO Contributes to Neurotoxicity in Alzheimer's Disease. J Neurosci 2021; 41:8461-8474. [PMID: 34446574 PMCID: PMC8513707 DOI: 10.1523/jneurosci.0494-21.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
α7 nicotinic acetylcholine receptors (nAChRs) are widely expressed in the brain where they promote fast cholinergic synaptic transmission and serve important neuromodulatory functions. However, their high permeability to Ca2+ also predisposes them to contribute to disease states. Here, using transfected HEK-tsa cells and primary cultured hippocampal neurons from male and female rats, we demonstrate that two proteins called Ly6h and NACHO compete for access to α7 subunits, operating together but in opposition to maintain α7 assembly and activity within a narrow range that is optimal for neuronal function and viability. Using mixed gender human temporal cortex and cultured hippocampal neurons from rats we further show that this balance is perturbed during Alzheimer's disease (AD) because of amyloid β (Aβ)-driven reduction in Ly6h, with severe reduction leading to increased phosphorylated tau and α7-mediated neurotoxicity. Ly6h release into human CSF is also correlated with AD severity. Thus, Ly6h links cholinergic signaling, Aβ and phosphorylated tau and may serve as a novel marker for AD progression.SIGNIFICANCE STATEMENT One of the earliest and most persistent hypotheses regarding Alzheimer's disease (AD) attributes cognitive impairment to loss of cholinergic signaling. More recently, interest has focused on crucial roles for amyloid β (Aβ) and phosphorylated tau in Alzheimer's pathogenesis. Here, we demonstrate that these elements are linked by Ly6h and its counterpart, NACHO, functioning in opposition to maintain assembly of nicotinic acetylcholine receptors (nAChRs) within the physiological range. Our data suggests that Aβ shifts the balance away from Ly6h and toward NACHO, resulting in increased assembly of Ca2+-permeable nAChRs and thus a conversion of basal cholinergic to neurotoxic signaling.
Collapse
Affiliation(s)
- Meilin Wu
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Clifford Z Liu
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Erika A Barrall
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, California 92093
- Alzheimer's Disease Research Center, University of California San Diego, La Jolla, California 92093
| | - William J Joiner
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
- Center for Circadian Biology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
30
|
Cheng Q, Lamb P, Stevanovic K, Bernstein BJ, Fry SA, Cushman JD, Yakel JL. Differential signalling induced by α7 nicotinic acetylcholine receptors in hippocampal dentate gyrus in vitro and in vivo. J Physiol 2021; 599:4687-4704. [PMID: 34487349 DOI: 10.1113/jp280505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/03/2021] [Indexed: 11/08/2022] Open
Abstract
The activation of α7 nicotinic acetylcholine receptors (nAChRs) has been shown to improve hippocampus-dependent learning and memory. α7 nAChRs are densely expressed among several different cell types in the hippocampus, with high Ca2+ permeability, although it is unclear if α7 nAChRs mobilize differential signalling mechanisms among distinct neuronal populations. To address this question, we compared α7 nAChR agonist-induced responses (i.e. calcium and cAMP changes) between granule cells and GABAergic neurons in the hippocampal dentate gyrus both in vitro and in vivo. In cultured organotypic hippocampal slices, we observed robust intracellular calcium and cAMP increases in dentate granule cells upon activation of α7 nAChRs. In contrast, GABAergic interneurons displayed little change in either calcium or cAMP concentration after α7 nAChR activation, even though they displayed much larger α7 nAChR current responses than those of dentate granule cells. We found that this was due to smaller α7 nAChR-induced Ca2+ rises in GABAergic interneurons. Thus, the regulation of the Ca2+ transients in different cell types resulted in differential subsequent intracellular signalling cascades and likely the ultimate outcome of α7 nAChR activation. Furthermore, we monitored neuronal activities of dentate granule cells and GABAergic interneurons in vivo via optic fibre photometry. We observed enhancement of neuronal activities after nicotine administration in dentate granule cells, but not in GABAergic neurons, which was absent in α7 nAChR-deficient granule cells. In summary, we reveal a mechanism for α7 nAChR-mediated increase of neuronal activity via cell type-specific intracellular signalling pathways. KEY POINTS: α7 nicotinic acetylcholine receptors (nAChRs) are widely distributed throughout the central nervous system and regulate a variety of brain functions including learning and memory. Understanding the cellular signalling mechanisms of their activations among different neuronal populations is important for delineating their actions in cognitive function, and developing effective treatment strategies for cognitive deficits. We report that α7 nAChR activation leads to Ca2+ and cAMP increases in granule cells (but not in GABAergic interneurons) in hippocampal dentate gyrus in vitro, a key region for pattern separation during learning. We also found that nicotine enhanced granule cell (but not in GABAergic interneurons) activity in an α7 nAChR-dependent manner via in vivo fibre photometry recording. Based on our findings, we propose that differential responses to α7 nAChR activation between granule cells and GABAergic interneurons is responsible for the increase of excitation by α7 nAChR agonists in hippocampal circuits synergistically.
Collapse
Affiliation(s)
- Qing Cheng
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA.,Biological/Biomedical Research Institute, North Carolina Central University, Durham, NC, USA
| | - Patricia Lamb
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| | - Korey Stevanovic
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| | - Briana J Bernstein
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| | - Sydney A Fry
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| | - Jesse D Cushman
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| | - Jerrel L Yakel
- Neurobiology Laboratory, The National Institute of Environmental Health Sciences/National Institutes of Health, Durham, NC, USA
| |
Collapse
|
31
|
Matta JA, Gu S, Davini WB, Bredt DS. Nicotinic acetylcholine receptor redux: Discovery of accessories opens therapeutic vistas. Science 2021; 373:373/6556/eabg6539. [PMID: 34385370 DOI: 10.1126/science.abg6539] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The neurotransmitter acetylcholine (ACh) acts in part through a family of nicotinic ACh receptors (nAChRs), which mediate diverse physiological processes including muscle contraction, neurotransmission, and sensory transduction. Pharmacologically, nAChRs are responsible for tobacco addiction and are targeted by medicines for hypertension and dementia. Nicotinic AChRs were the first ion channels to be isolated. Recent studies have identified molecules that control nAChR biogenesis, trafficking, and function. These nAChR accessories include protein and chemical chaperones as well as auxiliary subunits. Whereas some factors act on many nAChRs, others are receptor specific. Discovery of these regulatory mechanisms is transforming nAChR research in cells and tissues ranging from central neurons to spinal ganglia to cochlear hair cells. Nicotinic AChR-specific accessories also enable drug discovery on high-confidence targets for psychiatric, neurological, and auditory disorders.
Collapse
Affiliation(s)
| | | | - Weston B Davini
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson & Johnson, San Diego, CA 92121, USA
| | - David S Bredt
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson & Johnson, San Diego, CA 92121, USA.
| |
Collapse
|
32
|
Abstract
The α7-type nicotinic acetylcholine receptor is one of the most unique and interesting of all the members of the cys-loop superfamily of ligand-gated ion channels. Since it was first identified initially as a binding site for α-bungarotoxin in mammalian brain and later as a functional homomeric receptor with relatively high calcium permeability, it has been pursued as a potential therapeutic target for numerous indications, from Alzheimer disease to asthma. In this review, we discuss the history and state of the art for targeting α7 receptors, beginning with subtype-selective agonists and the basic pharmacophore for the selective activation of α7 receptors. A key feature of α7 receptors is their rapid desensitization by standard "orthosteric" agonist, and we discuss insights into the conformational landscape of α7 receptors that has been gained by the development of ligands binding to allosteric sites. Some of these sites are targeted by positive allosteric modulators that have a wide range of effects on the activation profile of the receptors. Other sites are targeted by direct allosteric agonist or antagonists. We include a perspective on the potential importance of α7 receptors for metabotropic as well as ionotropic signaling. We outline the challenges that exist for future development of drugs to target this important receptor and approaches that may be considered to address those challenges. SIGNIFICANCE STATEMENT: The α7-type nicotinic acetylcholine receptor (nAChR) is acknowledged as a potentially important therapeutic target with functional properties associated with both ionotropic and metabotropic signaling. The functional properties of α7 nAChR can be regulated in diverse ways with the variety of orthosteric and allosteric ligands described in this review.
Collapse
Affiliation(s)
- Roger L Papke
- Departments of Pharmacology and Therapeutics (R.L.P) and Chemistry (N.A.H.), University of Florida, Gainesville, FL
| | - Nicole A Horenstein
- Departments of Pharmacology and Therapeutics (R.L.P) and Chemistry (N.A.H.), University of Florida, Gainesville, FL
| |
Collapse
|
33
|
Eadaim A, Hahm ET, Justice ED, Tsunoda S. Cholinergic Synaptic Homeostasis Is Tuned by an NFAT-Mediated α7 nAChR-K v4/Shal Coupled Regulatory System. Cell Rep 2021; 32:108119. [PMID: 32905767 PMCID: PMC7521586 DOI: 10.1016/j.celrep.2020.108119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/23/2020] [Accepted: 08/17/2020] [Indexed: 11/26/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) involves compensatory mechanisms employed by neurons and circuits to preserve signaling when confronted with global changes in activity that may occur during physiological and pathological conditions. Cholinergic neurons, which are especially affected in some pathologies, have recently been shown to exhibit HSP mediated by nicotinic acetylcholine receptors (nAChRs). In Drosophila central neurons, pharmacological blockade of activity induces a homeostatic response mediated by the Drosophila α7 (Dα7) nAChR, which is tuned by a subsequent increase in expression of the voltage-dependent Kv4/Shal channel. Here, we show that an in vivo reduction of cholinergic signaling induces HSP mediated by Dα7 nAChRs, and this upregulation of Dα7 itself is sufficient to trigger transcriptional activation, mediated by nuclear factor of activated T cells (NFAT), of the Kv4/Shal gene, revealing a receptor-ion channel system coupled for homeostatic tuning in cholinergic neurons. Eadaim et al. show that in vivo reduction of cholinergic signaling in Drosophila neurons induces synaptic homeostasis mediated by Dα7 nAChRs. This upregulation of Dα7 induces Kv4/Shal gene expression mediated by nuclear factor of activated T cells (NFAT), revealing a receptor-ion channel system coupled for homeostatic tuning in cholinergic neurons.
Collapse
Affiliation(s)
- Abdunaser Eadaim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Eu-Teum Hahm
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Elizabeth D Justice
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
34
|
Borkar NA, Roos B, Prakash YS, Sathish V, Pabelick CM. Nicotinic α7 acetylcholine receptor (α7nAChR) in human airway smooth muscle. Arch Biochem Biophys 2021; 706:108897. [PMID: 34004182 DOI: 10.1016/j.abb.2021.108897] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/31/2022]
Abstract
Diseases such as asthma are exacerbated by inflammation, cigarette smoke and even nicotine delivery devices such as e-cigarettes. However, there is currently little information on how nicotine affects airways, particularly in humans, and changes in the context of inflammation or asthma. Here, a longstanding assumption is that airway smooth muscle (ASM) that is key to bronchoconstriction has muscarinic receptors while nicotinic receptors (nAChRs) are only on airway neurons. In this study, we tested the hypothesis that human ASM expresses α7nAChR and explored its profile in inflammation and asthma using ASM of non-asthmatics vs. mild-moderate asthmatics. mRNA and western analysis showed the α7 subunit is most expressed in ASM cells and further increased in asthmatics and smokers, or by exposure to nicotine, cigarette smoke or pro-inflammatory cytokines TNFα and IL-13. In these effects, signaling pathways relevant to asthma such as NFκB, AP-1 and CREB are involved. These novel data demonstrate the expression of α7nAChR in human ASM and suggest their potential role in asthma pathophysiology in the context of nicotine exposure.
Collapse
Affiliation(s)
- Niyati A Borkar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Benjamin Roos
- Department of Anesthesiology and Perioperative Medicine, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Sherafat Y, Bautista M, Fowler CD. Multidimensional Intersection of Nicotine, Gene Expression, and Behavior. Front Behav Neurosci 2021; 15:649129. [PMID: 33828466 PMCID: PMC8019722 DOI: 10.3389/fnbeh.2021.649129] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
The cholinergic system plays a crucial role in nervous system function with important effects on developmental processes, cognition, attention, motivation, reward, learning, and memory. Nicotine, the reinforcing component of tobacco and e-cigarettes, directly acts on the cholinergic system by targeting nicotinic acetylcholine receptors (nAChRs) in the brain. Activation of nAChRs leads to a multitude of immediate and long-lasting effects in specific cellular populations, thereby affecting the addictive properties of the drug. In addition to the direct actions of nicotine in binding to and opening nAChRs, the subsequent activation of circuits and downstream signaling cascades leads to a wide range of changes in gene expression, which can subsequently alter further behavioral expression. In this review, we provide an overview of the actions of nicotine that lead to changes in gene expression and further highlight evidence supporting how these changes can often be bidirectional, thereby inducing subsequent changes in behaviors associated with further drug intake.
Collapse
Affiliation(s)
- Yasmine Sherafat
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, Unites States
| | - Malia Bautista
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, Unites States
| | - Christie D Fowler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, Unites States
| |
Collapse
|
36
|
Marcovich I, Moglie MJ, Carpaneto Freixas AE, Trigila AP, Franchini LF, Plazas PV, Lipovsek M, Elgoyhen AB. Distinct Evolutionary Trajectories of Neuronal and Hair Cell Nicotinic Acetylcholine Receptors. Mol Biol Evol 2021; 37:1070-1089. [PMID: 31821508 PMCID: PMC7086180 DOI: 10.1093/molbev/msz290] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The expansion and pruning of ion channel families has played a crucial role in the evolution of nervous systems. Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels with distinct roles in synaptic transmission at the neuromuscular junction, the central and peripheral nervous system, and the inner ear. Remarkably, the complement of nAChR subunits has been highly conserved along vertebrate phylogeny. To ask whether the different subtypes of receptors underwent different evolutionary trajectories, we performed a comprehensive analysis of vertebrate nAChRs coding sequences, mouse single-cell expression patterns, and comparative functional properties of receptors from three representative tetrapod species. We found significant differences between hair cell and neuronal receptors that were most likely shaped by the differences in coexpression patterns and coassembly rules of component subunits. Thus, neuronal nAChRs showed high degree of coding sequence conservation, coupled to greater coexpression variance and conservation of functional properties across tetrapod clades. In contrast, hair cell α9α10 nAChRs exhibited greater sequence divergence, narrow coexpression pattern, and great variability of functional properties across species. These results point to differential substrates for random change within the family of gene paralogs that relate to the segregated roles of nAChRs in synaptic transmission.
Collapse
Affiliation(s)
- Irina Marcovich
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marcelo J Moglie
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Agustín E Carpaneto Freixas
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Anabella P Trigila
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lucia F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paola V Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcela Lipovsek
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Centre for Developmental Neurobiology, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Guy's Campus, London, United Kingdom
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
37
|
Structure and gating mechanism of the α7 nicotinic acetylcholine receptor. Cell 2021; 184:2121-2134.e13. [PMID: 33735609 DOI: 10.1016/j.cell.2021.02.049] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
The α7 nicotinic acetylcholine receptor plays critical roles in the central nervous system and in the cholinergic inflammatory pathway. This ligand-gated ion channel assembles as a homopentamer, is exceptionally permeable to Ca2+, and desensitizes faster than any other Cys-loop receptor. The α7 receptor has served as a prototype for the Cys-loop superfamily yet has proven refractory to structural analysis. We present cryo-EM structures of the human α7 nicotinic receptor in a lipidic environment in resting, activated, and desensitized states, illuminating the principal steps in the gating cycle. The structures also reveal elements that contribute to its function, including a C-terminal latch that is permissive for channel opening, and an anionic ring in the extracellular vestibule that contributes to its high conductance and calcium permeability. Comparisons among the α7 structures provide a foundation for mapping the gating cycle and reveal divergence in gating mechanisms in the Cys-loop receptor superfamily.
Collapse
|
38
|
Knowland D, Gu S, Eckert WA, Dawe GB, Matta JA, Limberis J, Wickenden AD, Bhattacharya A, Bredt DS. Functional α6β4 acetylcholine receptor expression enables pharmacological testing of nicotinic agonists with analgesic properties. J Clin Invest 2021; 130:6158-6170. [PMID: 33074244 DOI: 10.1172/jci140311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2020] [Indexed: 01/25/2023] Open
Abstract
The α6β4 nicotinic acetylcholine receptor (nAChR) is enriched in dorsal root ganglia neurons and is an attractive non-opioid therapeutic target for pain. However, difficulty expressing human α6β4 receptors in recombinant systems has precluded drug discovery. Here, genome-wide screening identified accessory proteins that enable reconstitution of human α6β4 nAChRs. BARP, an auxiliary subunit of voltage-dependent calcium channels, promoted α6β4 surface expression while IRE1α, an unfolded protein response sensor, enhanced α6β4 receptor assembly. Effects on α6β4 involve BARP's N-terminal region and IRE1α's splicing of XBP1 mRNA. Furthermore, clinical efficacy of nicotinic agents in relieving neuropathic pain best correlated with their activity on α6β4. Finally, BARP-knockout, but not NACHO-knockout mice lacked nicotine-induced antiallodynia, highlighting the functional importance of α6β4 in pain. These results identify roles for IRE1α and BARP in neurotransmitter receptor assembly and unlock drug discovery for the previously elusive α6β4 receptor.
Collapse
|
39
|
Grant S, Lester HA. Proteins for increased surface expression of the α6β4 nicotinic acetylcholine receptor: nothing but good news? J Clin Invest 2021; 130:5685-5687. [PMID: 33074245 DOI: 10.1172/jci143197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Useful animal models of disease in neuroscience can make accurate predictions about a therapeutic outcome, a feature known as predictive validity. In this issue of the JCI, Knowland et al. provide an improved model to assess nicotinic acetylcholine receptor (nAChR) ligands for treating chronic pain. The authors identify two proteins, the voltage-dependent calcium channel auxiliary subunit BARP and the unfolded protein response sensor IRE1α, that are required for robust heterologous expression of α6β4, an nAChR subtype in dorsal root ganglia (DRG). This nAChR is a candidate for the analgesic effects of nicotine as well as the frog toxin epibatidine. Now researchers can efficiently screen for α6β4 nAChR-selective agonists using heterologous expression systems. Candidates that emerge will enable researchers to test the predictive validity of mouse models for chronic pain in the nAChR context. If all these steps work, one can envision a class of non-opioid nAChR-targeted analgesics for chronic pain.
Collapse
Affiliation(s)
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
40
|
Mazzaferro S, Whiteman ST, Alcaino C, Beyder A, Sine SM. NACHO and 14-3-3 promote expression of distinct subunit stoichiometries of the α4β2 acetylcholine receptor. Cell Mol Life Sci 2021; 78:1565-1575. [PMID: 32676916 PMCID: PMC7854996 DOI: 10.1007/s00018-020-03592-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) belong to the superfamily of pentameric ligand-gated ion channels, and in neuronal tissues, are assembled from various types of α- and β-subunits. Furthermore, the subunits α4 and β2 assemble in two predominant stoichiometric forms, (α4)2(β2)3 and (α4)3(β2)2, forming receptors with dramatically different sensitivity to agonists and allosteric modulators. However, mechanisms by which the two stoichiometric forms are regulated are not known. Here, using heterologous expression in mammalian cells, single-channel patch-clamp electrophysiology, and calcium imaging, we show that the ER-resident protein NACHO selectively promotes the expression of the (α4)2(β2)3 stoichiometry, whereas the cytosolic molecular chaperone 14-3-3η selectively promotes the expression of the (α4)3(β2)2 stoichiometry. Thus, NACHO and 14-3-3η are potential physiological regulators of subunit stoichiometry, and are potential drug targets for re-balancing the stoichiometry in pathological conditions involving α4β2 nAChRs such as nicotine dependence and epilepsy.
Collapse
Affiliation(s)
- Simone Mazzaferro
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| | - Sara T Whiteman
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Constanza Alcaino
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Steven M Sine
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| |
Collapse
|
41
|
Han W, Shepard RD, Lu W. Regulation of GABA ARs by Transmembrane Accessory Proteins. Trends Neurosci 2021; 44:152-165. [PMID: 33234346 PMCID: PMC7855156 DOI: 10.1016/j.tins.2020.10.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
The vast majority of fast inhibitory transmission in the brain is mediated by GABA acting on GABAA receptors (GABAARs), which provides inhibitory balance to excitatory drive and controls neuronal output. GABAARs are also effectively targeted by clinically important drugs for treatment in a number of neurological disorders. It has long been hypothesized that function and pharmacology of GABAARs are determined by the channel pore-forming subunits. However, recent studies have provided new dimensions in studying GABAARs due to several transmembrane proteins that interact with GABAARs and modulate their trafficking and function. In this review, we summarize recent findings on these novel GABAAR transmembrane regulators and highlight a potential avenue to develop new GABAAR psychopharmacology by targeting these receptor-associated membrane proteins.
Collapse
Affiliation(s)
- Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan D Shepard
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
The nAChR Chaperone TMEM35a (NACHO) Contributes to the Development of Hyperalgesia in Mice. Neuroscience 2021; 457:74-87. [PMID: 33422618 PMCID: PMC7897319 DOI: 10.1016/j.neuroscience.2020.12.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 01/21/2023]
Abstract
Pain is a major health problem, affecting over fifty million adults in the US alone, with significant economic cost in medical care and lost productivity. Despite evidence implicating nicotinic acetylcholine receptors (nAChRs) in pathological pain, their specific contribution to pain processing in the spinal cord remains unclear given their presence in both neuronal and non-neuronal cell types. Here we investigated if loss of neuronal-specific TMEM35a (NACHO), a novel chaperone for functional expression of the homomeric α7 and assembly of the heteromeric α3, α4, and α6-containing nAChRs, modulates pain in mice. Mice with tmem35a deletion exhibited thermal hyperalgesia and mechanical allodynia. Intrathecal administration of nicotine and the α7-specific agonist, PHA543613, produced analgesic responses to noxious heat and mechanical stimuli in tmem35a KO mice, respectively, suggesting residual expression of these receptors or off-target effects. Since NACHO is expressed only in neurons, these findings indicate that neuronal α7 nAChR in the spinal cord contributes to heat nociception. To further determine the molecular basis underlying the pain phenotype, we analyzed the spinal cord transcriptome. Compared to WT control, the spinal cord of tmem35a KO mice exhibited 72 differentially-expressed genes (DEGs). These DEGs were mapped onto functional gene networks using the knowledge-based database, Ingenuity Pathway Analysis, and suggests increased neuroinflammation as a potential contributing factor for the hyperalgesia in tmem35a KO mice. Collectively, these findings implicate a heightened inflammatory response in the absence of neuronal NACHO activity. Additional studies are needed to determine the precise mechanism by which NACHO in the spinal cord modulates pain.
Collapse
|
43
|
Mesoy SM, Lummis SCR. M4, the Outermost Helix, is Extensively Involved in Opening of the α4β2 nACh Receptor. ACS Chem Neurosci 2021; 12:133-139. [PMID: 33295751 DOI: 10.1021/acschemneuro.0c00618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChR) are the archetypal members of the pentameric ligand-gated ion channel (pLGIC) family, an important class of cell signaling proteins. In all members of this family, each of the five subunits has four transmembrane α-helices (M1-M4), with M2 lining the pore, then M1 and M3, and with M4 outermost and adjacent to the membrane lipids. Despite its remote location, M4 contributes both to receptor assembly and gating in pLGICs where it has been examined. This study probes the role of M4 residues in the α4β2 nAChR using site-directed mutagenesis to individually mutate each residue to alanine, followed by expression in HEK293 cells and then characterization using membrane potential sensitive dye and radioligand binding. Two of the resulting mutant receptors showed altered EC50s, while 13 were nonfunctional, although coexpression with the chaperones RIC3 and nAChO resulted in 4 of these responding to agonist. Of the remaining 9, radioligand binding with epibatidine showed that 8 were expressed, suggesting these residues may play a role in channel opening. These data differ from similar studies in other pLGIC, where few or no Ala mutants in M4 ablate function, and they suggest that the α4β2 nAChR M4 may play a more significant role than in related receptors.
Collapse
Affiliation(s)
- Susanne M. Mesoy
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB 1QW, United Kingdom
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB 1QW, United Kingdom
| |
Collapse
|
44
|
Scholze P, Huck S. The α5 Nicotinic Acetylcholine Receptor Subunit Differentially Modulates α4β2 * and α3β4 * Receptors. Front Synaptic Neurosci 2020; 12:607959. [PMID: 33343327 PMCID: PMC7744819 DOI: 10.3389/fnsyn.2020.607959] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/11/2020] [Indexed: 11/30/2022] Open
Abstract
Nicotine, the principal reinforcing compound in tobacco, acts in the brain by activating neuronal nicotinic acetylcholine receptors (nAChRs). This review summarizes our current knowledge regarding how the α5 accessory nAChR subunit, encoded by the CHRNA5 gene, differentially modulates α4β2* and α3β4* receptors at the cellular level. Genome-wide association studies have linked a gene cluster in chromosomal region 15q25 to increased susceptibility to nicotine addiction, lung cancer, chronic obstructive pulmonary disease, and peripheral arterial disease. Interestingly, this gene cluster contains a non-synonymous single-nucleotide polymorphism (SNP) in the human CHRNA5 gene, causing an aspartic acid (D) to asparagine (N) substitution at amino acid position 398 in the α5 nAChR subunit. Although other SNPs have been associated with tobacco smoking behavior, efforts have focused predominantly on the D398 and N398 variants in the α5 subunit. In recent years, significant progress has been made toward understanding the role that the α5 nAChR subunit—and the role of the D398 and N398 variants—plays on nAChR function at the cellular level. These insights stem primarily from a wide range of experimental models, including receptors expressed heterologously in Xenopus oocytes, various cell lines, and neurons derived from human induced pluripotent stem cells (iPSCs), as well as endogenous receptors in genetically engineered mice and—more recently—rats. Despite providing a wealth of available data, however, these studies have yielded conflicting results, and our understanding of the modulatory role that the α5 subunit plays remains incomplete. Here, we review these reports and the various techniques used for expression and analysis in order to examine how the α5 subunit modulates key functions in α4β2* and α3β4* receptors, including receptor trafficking, sensitivity, efficacy, and desensitization. In addition, we highlight the strikingly different role that the α5 subunit plays in Ca2+ signaling between α4β2* and α3β4* receptors, and we discuss whether the N398 α5 subunit variant can partially replace the D398 variant.
Collapse
Affiliation(s)
- Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Sigismund Huck
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Miller DR, Khoshbouei H, Garai S, Cantwell LN, Stokes C, Thakur G, Papke RL. Allosterically Potentiated α7 Nicotinic Acetylcholine Receptors: Reduced Calcium Permeability and Current-Independent Control of Intracellular Calcium. Mol Pharmacol 2020; 98:695-709. [PMID: 33020143 PMCID: PMC7662531 DOI: 10.1124/molpharm.120.000012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/10/2020] [Indexed: 11/22/2022] Open
Abstract
The currents of α7 nicotinic acetylcholine receptors activated by acetylcholine (ACh) are brief. The channel has high permeability to calcium relative to monovalent cations and shows inward rectification. It has been previously noted that in the presence of positive allosteric modulators (PAMs), currents through the channels of α7 receptors differ from normal α7 currents both in sensitivity to specific channel blockers and their current-voltage (I-V) relationships, no longer showing inward rectification. Linear I-V functions are often associated with channels lacking calcium permeability, so we measured the I-V functions of α7 receptors activated by ACh when PAMs were bound to the allosteric binding site in the transmembrane domain. Currents were recorded in chloride-free Ringer's solution with low or high concentrations of extracellular calcium to determine the magnitude of the reversal potential shift in the two conditions as well as the I-V relationships. ACh-evoked currents potentiated by the allosteric agonist-PAMs (ago-PAMs) (3aR,4S,9bS)-4-(4-bromophenyl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline-8-sulfonamide (GAT107) and 3-(3,4-difluorophenyl)-N-(1-(6-(4-(pyridin-2-yl)piperazin-1-yl)pyrazin-2-yl)ethyl)propenamide (B-973B) showed reduced inward rectification and calcium-dependent reversal potential shifts decreased by 80%, and 50%, respectively, compared with currents activated by ACh alone, indicative of reduced calcium permeability. Currents potentiated by 3a,4,5,9b-tetrahydro-4-(1-naphthalenyl)-3H-cyclopentan[c]quinoline-8-sulfonamide were also linear and showed no calcium-dependent reversal potential shifts. The ago-PAMs GAT-107 and B-973B stimulated increases in intracellular calcium in stably transfected HEK293 cells. However, these calcium signals were delayed relative to channel activation produced by these agents and were insensitive to the channel blocker mecamylamine. Our results indicate that, although allosterically activated α7 nicotinic ACh receptor may affect intracellular calcium levels, such effects are not likely due to large channel-dependent calcium influx. SIGNIFICANCE STATEMENT: Positive allosteric modulators (PAMs) of α7 nicotinic acetylcholine receptor can increase channel activation by two or more orders of magnitude, raising the concern that, due to the relatively high calcium permeability of α7 receptors activated by acetylcholine alone, such efficacious PAMs may have cytotoxic side effects. We show that PAMs alter the ion conduction pathway and, in general, reduce the calcium permeability of the channels. This supports the hypothesis that α7 effects on intracellular calcium may be independent of channel-mediated calcium influx.
Collapse
Affiliation(s)
- Douglas R Miller
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Habibeh Khoshbouei
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Sumanta Garai
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Lucas N Cantwell
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Clare Stokes
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Ganesh Thakur
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| | - Roger L Papke
- Departments of Neuroscience (D.R.M., H.K.) and Pharmacology and Therapeutics (C.S., R.L.P.), University of Florida, Gainesville, Florida; and Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts (S.G., L.N.C., G.T.)
| |
Collapse
|
46
|
Implications of Oligomeric Amyloid-Beta (oAβ 42) Signaling through α7β2-Nicotinic Acetylcholine Receptors (nAChRs) on Basal Forebrain Cholinergic Neuronal Intrinsic Excitability and Cognitive Decline. J Neurosci 2020; 41:555-575. [PMID: 33239400 DOI: 10.1523/jneurosci.0876-20.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 01/08/2023] Open
Abstract
Neuronal and network-level hyperexcitability is commonly associated with increased levels of amyloid-β (Aβ) and contribute to cognitive deficits associated with Alzheimer's disease (AD). However, the mechanistic complexity underlying the selective loss of basal forebrain cholinergic neurons (BFCNs), a well-recognized characteristic of AD, remains poorly understood. In this study, we tested the hypothesis that the oligomeric form of amyloid-β (oAβ42), interacting with α7-containing nicotinic acetylcholine receptor (nAChR) subtypes, leads to subnucleus-specific alterations in BFCN excitability and impaired cognition. We used single-channel electrophysiology to show that oAβ42 activates both homomeric α7- and heteromeric α7β2-nAChR subtypes while preferentially enhancing α7β2-nAChR open-dwell times. Organotypic slice cultures were prepared from male and female ChAT-EGFP mice, and current-clamp recordings obtained from BFCNs chronically exposed to pathophysiologically relevant level of oAβ42 showed enhanced neuronal intrinsic excitability and action potential firing rates. These resulted from a reduction in action potential afterhyperpolarization and alterations in the maximal rates of voltage change during spike depolarization and repolarization. These effects were observed in BFCNs from the medial septum diagonal band and horizontal diagonal band, but not the nucleus basalis. Last, aged male and female APP/PS1 transgenic mice, genetically null for the β2 nAChR subunit gene, showed improved spatial reference memory compared with APP/PS1 aged-matched littermates. Combined, these data provide a molecular mechanism supporting a role for α7β2-nAChR in mediating the effects of oAβ42 on excitability of specific populations of cholinergic neurons and provide a framework for understanding the role of α7β2-nAChR in oAβ42-induced cognitive decline.
Collapse
|
47
|
Hair cell α9α10 nicotinic acetylcholine receptor functional expression regulated by ligand binding and deafness gene products. Proc Natl Acad Sci U S A 2020; 117:24534-24544. [PMID: 32929005 DOI: 10.1073/pnas.2013762117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Auditory hair cells receive olivocochlear efferent innervation, which refines tonotopic mapping, improves sound discrimination, and mitigates acoustic trauma. The olivocochlear synapse involves α9α10 nicotinic acetylcholine receptors (nAChRs), which assemble in hair cells only coincident with cholinergic innervation and do not express in recombinant mammalian cell lines. Here, genome-wide screening determined that assembly and surface expression of α9α10 require ligand binding. Ion channel function additionally demands an auxiliary subunit, which can be transmembrane inner ear (TMIE) or TMEM132e. Both of these single-pass transmembrane proteins are enriched in hair cells and underlie nonsyndromic human deafness. Inner hair cells from TMIE mutant mice show altered postsynaptic α9α10 function and retain α9α10-mediated transmission beyond the second postnatal week associated with abnormally persistent cholinergic innervation. Collectively, this study provides a mechanism to link cholinergic input with α9α10 assembly, identifies unexpected functions for human deafness genes TMIE/TMEM132e, and enables drug discovery for this elusive nAChR implicated in prevalent auditory disorders.
Collapse
|
48
|
Szigeti K, Ihnatovych I, Birkaya B, Chen Z, Ouf A, Indurthi DC, Bard JE, Kann J, Adams A, Chaves L, Sule N, Reisch JS, Pavlik V, Benedict RHB, Auerbach A, Wilding G. CHRFAM7A: A human specific fusion gene, accounts for the translational gap for cholinergic strategies in Alzheimer's disease. EBioMedicine 2020; 59:102892. [PMID: 32818803 PMCID: PMC7452451 DOI: 10.1016/j.ebiom.2020.102892] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/11/2020] [Accepted: 06/29/2020] [Indexed: 01/25/2023] Open
Abstract
Background Cholinergic neuronal loss is one of the hallmarks of AD related neurodegeneration; however, preclinical promise of α7 nAChR drugs failed to translate into humans. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of α7 nAChR and was unaccounted for in preclinical models. Methods Molecular methods: Function of CHRFAM7A alleles was studied in vitro in two disease relevant phenotypic readouts: electrophysiology and Aβ uptake. Genome edited human induced pluripotent stem cells (iPSC) were used as a model system with the human context. Double blind pharmacogenetic study: We performed double-blind pharmacogenetic analysis on the effect of AChEI therapy based on CHRFAM7A carrier status in two paradigms: response to drug initiation and DMT effect. Mini Mental Status Examination (MMSE) was used as outcome measure. Change in MMSE score from baseline was compared by 2-tailed T-test. Longitudinal analysis of clinical outcome (MMSE) was performed using a fitted general linear model, based on an assumed autoregressive covariance structure. Model independent variables included age, sex, and medication regimen at the time of the first utilized outcome measure (AChEI alone or AChEI plus memantine), APOE4 carrier status (0, 1 or 2 alleles as categorical variables) and CHRFAM7A genotype. Findings The direct and inverted alleles have distinct phenotypes. Functional CHRFAM7A allele classifies the population as 25% non-carriers and 75% carriers. Induced pluripotent stem cell (iPSC) models α7 nAChR mediated Aβ neurotoxicity. Pharmacological readout translates into both first exposure (p = 0.037) and disease modifying effect (p = 0.0048) in two double blind pharmacogenetic studies. Interpretation CHRFAM7A accounts for the translational gap in cholinergic strategies in AD. Clinical trials not accounting for this uniquely human genetic factor may have rejected drug candidates that would benefit 25% of AD. Reanalyses of the completed trials using this pharmacogenetic paradigm may identify effective therapy. Funding:
Collapse
Affiliation(s)
- Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| | - Ivanna Ihnatovych
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Barbara Birkaya
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ziqiang Chen
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Aya Ouf
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Dinesh C Indurthi
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Jonathan E Bard
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Julien Kann
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Alexandrea Adams
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Lee Chaves
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Norbert Sule
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY 14203, USA
| | - Joan S Reisch
- UT Southwestern, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Valory Pavlik
- Baylor College of Medicine, 1 Baylor Plz, Houston, TX 77030, USA
| | - Ralph H B Benedict
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Anthony Auerbach
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Gregory Wilding
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| |
Collapse
|
49
|
Chrna5 is Essential for a Rapid and Protected Response to Optogenetic Release of Endogenous Acetylcholine in Prefrontal Cortex. J Neurosci 2020; 40:7255-7268. [PMID: 32817066 DOI: 10.1523/jneurosci.1128-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/01/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022] Open
Abstract
Optimal attention performance requires cholinergic modulation of corticothalamic neurons in the prefrontal cortex. These pyramidal cells express specialized nicotinic acetylcholine receptors containing the α5 subunit encoded by Chrna5 Disruption of this gene impairs attention, but the advantage α5 confers on endogenous cholinergic signaling is unknown. To ascertain this underlying mechanism, we used optogenetics to stimulate cholinergic afferents in prefrontal cortex brain slices from compound-transgenic wild-type and Chrna5 knock-out mice of both sexes. These electrophysiological experiments identify that Chrna5 is critical for the rapid onset of the postsynaptic cholinergic response. Loss of α5 slows cholinergic excitation and delays its peak, and these effects are observed in two different optogenetic mouse lines. Disruption of Chrna5 does not otherwise perturb the magnitude of the response, which remains strongly mediated by nicotinic receptors and tightly controlled by autoinhibition via muscarinic M2 receptors. However, when conditions are altered to promote sustained cholinergic receptor stimulation, it becomes evident that α5 also works to protect nicotinic responses against desensitization. Rescuing Chrna5 disruption thus presents the double challenge of improving the onset of nicotinic signaling without triggering desensitization. Here, we identify that an agonist for the unorthodox α-α nicotinic binding site can allosterically enhance the cholinergic pathway considered vital for attention. Treatment with NS9283 restores the rapid onset of the postsynaptic cholinergic response without triggering desensitization. Together, this work demonstrates the advantages of speed and resilience that Chrna5 confers on endogenous cholinergic signaling, defining a critical window of interest for cue detection and attentional processing.SIGNIFICANCE STATEMENT The α5 nicotinic receptor subunit (Chrna5) is important for attention, but its advantage in detecting endogenous cholinergic signals is unknown. Here, we show that α5 subunits permit rapid cholinergic responses in prefrontal cortex and protect these responses from desensitization. Our findings clarify why Chrna5 is required for optimal attentional performance under demanding conditions. To treat the deficit arising from Chrna5 disruption without triggering desensitization, we enhanced nicotinic receptor affinity using NS9283 stimulation at the unorthodox α-α nicotinic binding site. This approach successfully restored the rapid-onset kinetics of endogenous cholinergic neurotransmission. In summary, we reveal a previously unknown role of Chrna5 as well as an effective approach to compensate for genetic disruption and permit fast cholinergic excitation of prefrontal attention circuits.
Collapse
|
50
|
NACHO Engages N-Glycosylation ER Chaperone Pathways for α7 Nicotinic Receptor Assembly. Cell Rep 2020; 32:108025. [DOI: 10.1016/j.celrep.2020.108025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/13/2020] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
|