α7- and α9-Containing Nicotinic Acetylcholine Receptors in the Functioning of Immune System and in Pain

Nicotinic acetylcholine receptors (nAChRs) present as many different subtypes in the nervous and immune systems, muscles and on the cells of other organs. In the immune system, inflammation is regulated via the vagus nerve through the activation of the non-neuronal α7 nAChR subtype, affecting the production of cytokines. The analgesic properties of α7 nAChR-selective compounds are mostly based on the activation of the cholinergic anti-inflammatory pathway. The molecular mechanism of neuropathic pain relief mediated by the inhibition of α9-containing nAChRs is not fully understood yet, but the role of immune factors in this process is becoming evident. To obtain appropriate drugs, a search of selective agonists, antagonists and modulators of α7- and α9-containing nAChRs is underway. The naturally occurring three-finger snake α-neurotoxins and mammalian Ly6/uPAR proteins, as well as neurotoxic peptides α-conotoxins, are not only sophisticated tools in research on nAChRs but are also considered as potential medicines. In particular, the inhibition of the α9-containing nAChRs by α-conotoxins may be a pathway to alleviate neuropathic pain. nAChRs are involved in the inflammation processes during AIDS and other viral infections; thus they can also be means used in drug design. In this review, we discuss the role of α7- and α9-containing nAChRs in the immune processes and in pain.

[1]  M. Lotz,et al.  Human-specific duplicate CHRFAM7A gene is associated with more severe osteoarthritis and amplifies pain behaviours , 2023, Annals of the Rheumatic Diseases.

[2]  Ping Zou,et al.  Up‐regulation of the human‐specific CHRFAM7A gene protects against renal fibrosis in mice with obstructive nephropathy , 2022, Journal of cellular and molecular medicine.

[3]  C. Ghelardini,et al.  marine drugs , 2022 .

[4]  D. Nolde,et al.  Membrane-mediated interaction of non-conventional snake three-finger toxins with nicotinic acetylcholine receptors , 2022, Communications Biology.

[5]  J. McIntosh,et al.  RgIA4 Prevention of Acute Oxaliplatin-Induced Cold Allodynia Requires α9-Containing Nicotinic Acetylcholine Receptors and CD3+ T-Cells , 2022, Cells.

[6]  G. Cymes,et al.  An experimental test of the nicotinic hypothesis of COVID-19 , 2022, Proceedings of the National Academy of Sciences of the United States of America.

[7]  X. Tian,et al.  Targeting α7 nicotinic acetylcholine receptors for chronic pain , 2022, Frontiers in Molecular Neuroscience.

[8]  David John Adams,et al.  Mechanism of Action and Structure-Activity Relationship of α-Conotoxin Mr1.1 at the Human α9α10 Nicotinic Acetylcholine Receptor. , 2022, Journal of medicinal chemistry.

[9]  Chenyun Guo,et al.  Discovery and Structural and Functional Characterization of a Novel A-Superfamily Conotoxin Targeting α9α10 Nicotinic Acetylcholine Receptor. , 2022, ACS chemical biology.

[10]  K. Candiotti,et al.  Mechanism of Electroacupuncture Analgesia on Nicotine Withdrawal-Induced Hyperalgesia in a Rat Model , 2022, Evidence-based complementary and alternative medicine : eCAM.

[11]  J. Che,et al.  Novel αO-conotoxin GeXIVA[1,2] Nonaddictive Analgesic with Pharmacokinetic Modelling-Based Mechanistic Assessment , 2022, Pharmaceutics.

[12]  D. Bertrand,et al.  The molecular mechanism of snake short-chain α-neurotoxin binding to muscle-type nicotinic acetylcholine receptors , 2022, Nature Communications.

[13]  P. Ling,et al.  Tobacco product use and the risks of SARS-CoV-2 infection and COVID-19: current understanding and recommendations for future research , 2022, The Lancet Respiratory Medicine.

[14]  Xitong Dang,et al.  The emergence of the uniquely human α7 nicotinic acetylcholine receptor gene and its roles in inflammation. , 2022, Gene.

[15]  Jing Liu,et al.  Preparation of uniform-sized GeXIVA[1,2]-loaded PLGA microspheres as long-effective release system with high encapsulation efficiency , 2022, Drug delivery.

[16]  C. Bouzat,et al.  A Functional Interaction Between Y674-R685 Region of the SARS-CoV-2 Spike Protein and the Human α7 Nicotinic Receptor , 2022, Molecular Neurobiology.

[17]  M. Pallavicini,et al.  From 2-Triethylammonium Ethyl Ether of 4-Stilbenol (MG624) to Selective Small-Molecule Antagonists of Human α9α10 Nicotinic Receptor by Modifications at the Ammonium Ethyl Residue , 2022, Journal of medicinal chemistry.

[18]  K. Tracey,et al.  Manipulation of the inflammatory reflex as a therapeutic strategy , 2022, Cell reports. Medicine.

[19]  S. Komisarenko,et al.  Immunization with 674–685 fragment of SARS-Cov-2 spike protein induces neuroinflammation and impairs episodic memory of mice , 2022, Biochemical and Biophysical Research Communications.

[20]  Matthew S. Jones,et al.  Letters to the editor: Nicotinic acetylcholine receptor ligands as potential targets for managing neuropathic pain induced by diabetic peripheral neuropathy , 2022, eNeurologicalSci.

[21]  Xiaoxuan Liu,et al.  Activation of Cholinergic Anti-Inflammatory Pathway Ameliorates Cerebral and Cardiac Dysfunction After Intracerebral Hemorrhage Through Autophagy , 2022, Frontiers in Immunology.

[22]  Dongting Zhangsun,et al.  Oligo-basic amino acids, potential nicotinic acetylcholine receptor inhibitors. , 2022, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[23]  F. Berenbaum,et al.  Alpha-7 Nicotinic Receptor Dampens Murine Osteoblastic Response to Inflammation and Age-Related Osteoarthritis , 2022, Frontiers in Immunology.

[24]  R. Papke,et al.  Comparison of the Anti-inflammatory Properties of Two Nicotinic Acetylcholine Receptor Ligands, Phosphocholine and pCF3-diEPP , 2022, Frontiers in Cellular Neuroscience.

[25]  D. Fornasari,et al.  The Human-Restricted Isoform of the α7 nAChR, CHRFAM7A: A Double-Edged Sword in Neurological and Inflammatory Disorders , 2022, International journal of molecular sciences.

[26]  T. Ma,et al.  The α7 Nicotinic Acetylcholine Receptor Agonist GTS-21 Improves Bacterial Clearance via Regulation of Monocyte Recruitment and Activity in Polymicrobial Septic Peritonitis , 2022, Frontiers in Immunology.

[27]  Y. Tan,et al.  Inflammation Regulation via an Agonist and Antagonists of α7 Nicotinic Acetylcholine Receptors in RAW264.7 Macrophages , 2022, Marine drugs.

[28]  R. Papke,et al.  Selective Agonists and Antagonists of α9 Versus α7 Nicotinic Acetylcholine Receptors. , 2022, ACS chemical neuroscience.

[29]  J. Coleman,et al.  Structures of highly flexible intracellular domain of human α7 nicotinic acetylcholine receptor , 2022, Nature communications.

[30]  O. Kaminuma,et al.  Expression and Function of Nicotinic Acetylcholine Receptors in Induced Regulatory T Cells , 2022, International journal of molecular sciences.

[31]  Hong Liu,et al.  Role of Cholinergic Anti-Inflammatory Pathway in Protecting Sepsis-Induced Acute Lung Injury through Regulation of the Conventional Dendritic Cells , 2022, Mediators of inflammation.

[32]  Y. Utkin,et al.  Marine Origin Ligands of Nicotinic Receptors: Low Molecular Compounds, Peptides and Proteins for Fundamental Research and Practical Applications , 2022, Biomolecules.

[33]  Y. Utkin,et al.  α-Conotoxin RgIA and oligoarginine R8 in the mice model alleviate long-term oxaliplatin induced neuropathy. , 2021, Biochimie.

[34]  E. Saphire,et al.  Structure of the rabies virus glycoprotein trimer bound to a prefusion-specific neutralizing antibody , 2021, bioRxiv.

[35]  D. Craik,et al.  ɑO‐Conotoxin GeXIVA isomers modulate N‐type calcium (CaV2.2) channels and inwardly‐rectifying potassium (GIRK) channels via GABAB receptor activation , 2021, Journal of Neurochemistry.

[36]  M. Munafo,et al.  Smoking and COVID-19 outcomes: an observational and Mendelian randomisation study using the UK Biobank cohort , 2021, Thorax.

[37]  R. Papke,et al.  Therapeutic Targeting of α7 Nicotinic Acetylcholine Receptors , 2021, Pharmacological Reviews.

[38]  Johnny J. He,et al.  Activation of α7 nicotinic acetylcholine receptor ameliorates HIV-associated neurology and neuropathology , 2021, Brain : a journal of neurology.

[39]  J. McIntosh,et al.  Discovery of Methylene Thioacetal-Incorporated α-RgIA Analogues as Potent and Stable Antagonists of the Human α9α10 Nicotinic Acetylcholine Receptor for the Treatment of Neuropathic Pain. , 2021, Journal of medicinal chemistry.

[40]  J. McIntosh,et al.  Selective Penicillamine Substitution Enables Development of a Potent Analgesic Peptide that Acts through a Non-Opioid-Based Mechanism. , 2021, Journal of medicinal chemistry.

[41]  David John Adams,et al.  Globular and ribbon isomers of Conus geographus α-conotoxins antagonize human nicotinic acetylcholine receptors. , 2021, Biochemical pharmacology.

[42]  D. Craik,et al.  Engineered Conotoxin Differentially Blocks and Discriminates Rat and Human α7 Nicotinic Acetylcholine Receptors. , 2021, Journal of medicinal chemistry.

[43]  S. Bonassi,et al.  Nicotine upregulates ACE2 expression and increases competence for SARS-CoV-2 in human pneumocytes , 2021, ERJ Open Research.

[44]  U. Gieler,et al.  New Pathways for the Skin's Stress Response: The Cholinergic Neuropeptide SLURP-1 Can Activate Mast Cells and Alter Cytokine Production in Mice , 2021, Frontiers in Immunology.

[45]  J. Gordon,et al.  Regulatory Dendritic Cells, T Cell Tolerance, and Dendritic Cell Therapy for Immunologic Disease , 2021, Frontiers in Immunology.

[46]  Hui Zhang,et al.  Lyophilization Serves as an Effective Strategy for Drug Development of the α9α10 Nicotinic Acetylcholine Receptor Antagonist α-Conotoxin GeXIVA[1,2] , 2021, Marine drugs.

[47]  T. Kuiken,et al.  Street RABV Induces the Cholinergic Anti-inflammatory Pathway in Human Monocyte-Derived Macrophages by Binding to nAChr α7 , 2021, Frontiers in Immunology.

[48]  Rommie E. Amaro,et al.  A potential interaction between the SARS-CoV-2 spike protein and nicotinic acetylcholine receptors , 2021, Biophysical Journal.

[49]  K. Farsalinos,et al.  Nicotinic cholinergic system and COVID-19: In silico identification of interactions between α7 nicotinic acetylcholine receptor and the cryptic epitopes of SARS-Co-V and SARS-CoV-2 Spike glycoproteins , 2021, Food and Chemical Toxicology.

[50]  O. Quesada,et al.  Disruption of the cholinergic anti-inflammatory response by R5-tropic HIV-1 protein gp120JRFL , 2021, The Journal of biological chemistry.

[51]  A. Chávez-Reyes,et al.  The human-specific duplicated α7 gene inhibits the ancestral α7, negatively regulating nicotinic acetylcholine receptor-mediated transmitter release , 2021, The Journal of biological chemistry.

[52]  V. Lemos,et al.  Role of the α7 Nicotinic Acetylcholine Receptor in the Pathophysiology of Atherosclerosis , 2020, Frontiers in Physiology.

[53]  K. Mineev,et al.  Spatial Structure and Activity of Synthetic Fragments of Lynx1 and of Nicotinic Receptor Loop C Models , 2020, Biomolecules.

[54]  Yan-yan Chu,et al.  Medicinal chemistry, pharmacology, and therapeutic potential of α-conotoxins antagonizing the α9α10 nicotinic acetylcholine receptor. , 2020, Pharmacology & therapeutics.

[55]  I. Tikhonova,et al.  α9α10 nicotinic acetylcholine receptors regulate murine bone marrow granulocyte functions. , 2020, Immunobiology.

[56]  R. Lewis,et al.  Structure-Function of Neuronal Nicotinic Acetylcholine Receptor Inhibitors Derived From Natural Toxins , 2020, Frontiers in Neuroscience.

[57]  M. Kirpichnikov,et al.  Structural Diversity and Dynamics of Human Three-Finger Proteins Acting on Nicotinic Acetylcholine Receptors , 2020, International journal of molecular sciences.

[58]  Georgios E. Papadopoulos,et al.  Nicotinic Cholinergic System and COVID-19: In Silico Identification of an Interaction between SARS-CoV-2 and Nicotinic Receptors with Potential Therapeutic Targeting Implications , 2020, International journal of molecular sciences.

[59]  Y. Utkin,et al.  Three‐finger proteins from snakes and humans acting on nicotinic receptors: Old and new , 2020, Journal of neurochemistry.

[60]  M. Kloc,et al.  How nicotine can inhibit cytokine storm in the lungs and prevent or lessen the severity of COVID-19 infection? , 2020, Immunology Letters.

[61]  J. McIntosh,et al.  Behavioral and Molecular Basis of Cholinergic Modulation of Pain: Focus on Nicotinic Acetylcholine Receptors. , 2020, Current topics in behavioral neurosciences.

[62]  Y. Qadri,et al.  Implications for Neuromodulation Therapy to Control Inflammation and Related Organ Dysfunction in COVID-19 , 2020, Journal of Cardiovascular Translational Research.

[63]  M. Motaghinejad,et al.  Survival of COVID-19 patients requires precise immune regulation: The hypothetical immunoprotective role of nicotinic agonists , 2020, Medical Hypotheses.

[64]  K. Farsalinos,et al.  Systematic review of the prevalence of current smoking among hospitalized COVID-19 patients in China: could nicotine be a therapeutic option? , 2020, Internal and Emergency Medicine.

[65]  K. Sałat Chemotherapy-induced peripheral neuropathy—part 2: focus on the prevention of oxaliplatin-induced neurotoxicity , 2020, Pharmacological Reports.

[66]  Patrizia Russo,et al.  COVID-19 and smoking: is nicotine the hidden link? , 2020, European Respiratory Journal.

[67]  J. Changeux,et al.  A nicotinic hypothesis for Covid-19 with preventive and therapeutic implications. , 2020, Comptes rendus biologies.

[68]  A. Karlin,et al.  Structure of the Native Muscle-type Nicotinic Receptor and Inhibition by Snake Venom Toxins , 2020, Neuron.

[69]  P. Balaban,et al.  Water‐soluble variant of human Lynx1 positively modulates synaptic plasticity and ameliorates cognitive impairment associated with α7‐nAChR dysfunction , 2020, Journal of neurochemistry.

[70]  Y. Utkin,et al.  Activation of α7 Nicotinic Acetylcholine Receptor Upregulates HLA-DR and Macrophage Receptors: Potential Role in Adaptive Immunity and in Preventing Immunosuppression , 2020, Biomolecules.

[71]  M. Kirpichnikov,et al.  Human secreted protein SLURP-1 abolishes nicotine-induced proliferation, PTEN down-regulation and α7-nAChR expression up-regulation in lung cancer cells. , 2020, International immunopharmacology.

[72]  K. Kawashima,et al.  α7 nAChRs expressed on antigen presenting cells are insensitive to the conventional antagonists α-bungarotoxin and methyllycaconitine. , 2020, International Immunopharmacology.

[73]  J. McIntosh,et al.  RgIA4 Accelerates Recovery from Paclitaxel-Induced Neuropathic Pain in Rats , 2019, Marine drugs.

[74]  W. D. de Jonge,et al.  Acetylcholine-producing T-cells augment innate immune driven colitis but are redundant in T-cell driven colitis. , 2019, American journal of physiology. Gastrointestinal and liver physiology.

[75]  Jie Zhang,et al.  Migration of gastric cancer is suppressed by recombinant Newcastle disease virus (rL-RVG) via regulating α7-nicotinic acetylcholine receptors/ERK- EMT , 2019, BMC Cancer.

[76]  D. Craik,et al.  Conotoxins: Chemistry and Biology. , 2019, Chemical reviews.

[77]  J. Ecklund,et al.  Association of a Functional Polymorphism in the CHRFAM7A Gene with Inflammatory Response Mediators and Neuropathic Pain after Spinal Cord Injury. , 2019, Journal of neurotrauma.

[78]  N. Staff,et al.  Platinum‐induced peripheral neurotoxicity: From pathogenesis to treatment , 2019, Journal of the peripheral nervous system : JPNS.

[79]  Anil Tiwari,et al.  The secreted Ly-6/uPAR related protein-1 (SLURP1) stabilizes epithelial cell junctions and suppresses TNF-α-induced cytokine production. , 2019, Biochemical and biophysical research communications.

[80]  David John Adams,et al.  α-Conotoxin Vc1.1 Structure-Activity Relationship at the Human α9α10 Nicotinic Acetylcholine Receptor Investigated by Minimal Side Chain Replacement. , 2019, ACS chemical neuroscience.

[81]  Y. Utkin,et al.  From Synthetic Fragments of Endogenous Three-Finger Proteins to Potential Drugs , 2019, Front. Pharmacol..

[82]  D. Fornasari,et al.  Effect of donepezil on the expression and responsiveness to LPS of CHRNA7 and CHRFAM7A in macrophages: A possible link to the cholinergic anti-inflammatory pathway , 2019, Journal of Neuroimmunology.

[83]  K. Kawashima,et al.  Distinct Roles of α7 nAChRs in Antigen-Presenting Cells and CD4+ T Cells in the Regulation of T Cell Differentiation , 2019, Front. Immunol..

[84]  R. Su,et al.  The α9α10 Nicotinic Acetylcholine Receptor Antagonist αO-Conotoxin GeXIVA[1,2] Alleviates and Reverses Chemotherapy-Induced Neuropathic Pain , 2019, Marine drugs.

[85]  V. Tsetlin,et al.  Crystal Structure of the Monomeric Extracellular Domain of α9 Nicotinic Receptor Subunit in Complex With α-Conotoxin RgIA: Molecular Dynamics Insights Into RgIA Binding to α9α10 Nicotinic Receptors , 2019, Front. Pharmacol..

[86]  J. Miwa,et al.  Lynx Prototoxins: Roles of Endogenous Mammalian Neurotoxin-Like Proteins in Modulating Nicotinic Acetylcholine Receptor Function to Influence Complex Biological Processes , 2019, Front. Pharmacol..

[87]  G. Upadhyay Emerging Role of Lymphocyte Antigen-6 Family of Genes in Cancer and Immune Cells , 2019, Front. Immunol..

[88]  V. Jhanji,et al.  The secreted Ly6/uPAR-related protein-1 suppresses neutrophil binding, chemotaxis, and transmigration through human umbilical vein endothelial cells , 2019, Scientific Reports.

[89]  J. McIntosh,et al.  Conopeptides [V11L;V16D]ArIB and RgIA4: Powerful Tools for the Identification of Novel Nicotinic Acetylcholine Receptors in Monocytes , 2019, Front. Pharmacol..

[90]  T. Deer,et al.  Intrathecal Therapy for Chronic Pain: A Review of Morphine and Ziconotide as Firstline Options , 2018, Pain medicine.

[91]  R. Katsarava,et al.  Oligoarginine Peptides, a New Family of nAChR Inhibitors. , 2019, Molecular Pharmacology.

[92]  Hui Luo,et al.  The effect of the cholinergic anti-inflammatory pathway on collagen-induced arthritis involves the modulation of dendritic cell differentiation , 2018, Arthritis Research & Therapy.

[93]  Michael A. Faltys,et al.  Adenylyl Cyclase 6 Mediates Inhibition of TNF in the Inflammatory Reflex , 2018, Front. Immunol..

[94]  V. Tsetlin,et al.  Species specificity of rat and human α7 nicotinic acetylcholine receptors towards different classes of peptide and protein antagonists , 2018, Neuropharmacology.

[95]  R. Papke,et al.  The Antinociceptive and Anti-Inflammatory Properties of the α7 nAChR Weak Partial Agonist p-CF3 N,N-diethyl-N′-phenylpiperazine , 2018, The Journal of Pharmacology and Experimental Therapeutics.

[96]  W. Padberg,et al.  Phosphocholine-Modified Lipooligosaccharides of Haemophilus influenzae Inhibit ATP-Induced IL-1β Release by Pulmonary Epithelial Cells , 2018, Molecules.

[97]  I. König,et al.  C-Reactive Protein Stimulates Nicotinic Acetylcholine Receptors to Control ATP-Mediated Monocytic Inflammasome Activation , 2018, Front. Immunol..

[98]  J. Lasalde-Dominicci,et al.  Nicotinic Acetylcholine Receptors in HIV: Possible Roles During HAND and Inflammation , 2018, Cellular and Molecular Neurobiology.

[99]  W. Im,et al.  Augmenting the antinociceptive effects of nicotinic acetylcholine receptor activity through lynx1 modulation , 2018, PloS one.

[100]  R. Lewis,et al.  Neuronal Nicotinic Acetylcholine Receptor Modulators from Cone Snails , 2018, Marine drugs.

[101]  C. Enjalbal,et al.  Synthesis, Structure and Biological Activity of CIA and CIB, Two α-Conotoxins from the Predation-Evoked Venom of Conus catus , 2018, Toxins.

[102]  P. Dougherty,et al.  Beyond symptomatic relief for chemotherapy‐induced peripheral neuropathy: Targeting the source , 2018, Cancer.

[103]  D. Servent,et al.  α9‐containing nicotinic acetylcholine receptors and the modulation of pain , 2018, British journal of pharmacology.

[104]  M. Skok,et al.  α7 nicotinic acetylcholine receptors are involved in suppression of the antibody immune response , 2018, Journal of Neuroimmunology.

[105]  J. Lasalde-Dominicci,et al.  The Cholinergic Anti-Inflammatory Response and the Role of Macrophages in HIV-Induced Inflammation , 2018, International journal of molecular sciences.

[106]  R. Nichols,et al.  Beyond the Channel: Metabotropic Signaling by Nicotinic Receptors. , 2018, Trends in pharmacological sciences.

[107]  D. Dolgikh,et al.  Human secreted proteins SLURP‐1 and SLURP‐2 control the growth of epithelial cancer cells via interactions with nicotinic acetylcholine receptors , 2018, British journal of pharmacology.

[108]  K. Felix,et al.  Endogenous CHRNA7-ligand SLURP1 as a potential tumor suppressor and anti-nicotinic factor in pancreatic cancer , 2018, Oncotarget.

[109]  D. Craik,et al.  Interaction of Synthetic Human SLURP-1 with the Nicotinic Acetylcholine Receptors , 2017, Scientific Reports.

[110]  R. Papke,et al.  Rabies virus modifies host behaviour through a snake-toxin like region of its glycoprotein that inhibits neurotransmitter receptors in the CNS , 2017, Scientific Reports.

[111]  P. Keating,et al.  Effects of α-conotoxin ImI on TNF-α, IL-8 and TGF-β expression by human macrophage-like cells derived from THP-1 pre-monocytic leukemic cells , 2017, Scientific Reports.

[112]  K. Kawashima,et al.  Expression and Function of the Cholinergic System in Immune Cells , 2017, Front. Immunol..

[113]  Shuo Yu,et al.  Cloning, expression and functional characterization of a D-superfamily conotoxin Lt28.1 with previously undescribed cysteine pattern , 2017, Peptides.

[114]  E. Glowatzki,et al.  RgIA4 Potently Blocks Mouse α9α10 nAChRs and Provides Long Lasting Protection against Oxaliplatin-Induced Cold Allodynia , 2017, Front. Cell. Neurosci..

[115]  F. Shi,et al.  Acetylcholine-producing NK cells attenuate CNS inflammation via modulation of infiltrating monocytes/macrophages , 2017, Proceedings of the National Academy of Sciences.

[116]  W. Padberg,et al.  Canonical and Novel Non-Canonical Cholinergic Agonists Inhibit ATP-Induced Release of Monocytic Interleukin-1β via Different Combinations of Nicotinic Acetylcholine Receptor Subunits α7, α9 and α10 , 2017, Front. Cell. Neurosci..

[117]  C. Zhao,et al.  Monocytes primed with GTS-21/&agr;7 nAChR (nicotinic acetylcholine receptor) agonist develop anti-inflammatory memory , 2017, QJM : monthly journal of the Association of Physicians.

[118]  David John Adams,et al.  Identification of a Novel O-Conotoxin Reveals an Unusual and Potent Inhibitor of the Human α9α10 Nicotinic Acetylcholine Receptor , 2017, Marine drugs.

[119]  C. Ruppert,et al.  Surfactant inhibits ATP-induced release of interleukin-1β via nicotinic acetylcholine receptors[S] , 2017, Journal of Lipid Research.

[120]  K. Kawashima,et al.  Physiological functions of the cholinergic system in immune cells. , 2017, Journal of pharmacological sciences.

[121]  C. Ghelardini,et al.  Inhibition of α9α10 nicotinic acetylcholine receptors prevents chemotherapy-induced neuropathic pain , 2017, Proceedings of the National Academy of Sciences.

[122]  S. Im,et al.  Incidence of taxane-induced peripheral neuropathy receiving treatment and prescription patterns in patients with breast cancer , 2017, Supportive Care in Cancer.

[123]  A. Ceballos,et al.  Acetylcholine polarizes dendritic cells toward a Th2‐promoting profile , 2017, Allergy.

[124]  N. Knuckey,et al.  The Neuroprotective Peptide Poly-Arginine-12 (R12) Reduces Cell Surface Levels of NMDA NR2B Receptor Subunit in Cortical Neurons; Investigation into the Involvement of Endocytic Mechanisms , 2016, Journal of Molecular Neuroscience.

[125]  R. Efremov,et al.  High-Affinity α-Conotoxin PnIA Analogs Designed on the Basis of the Protein Surface Topography Method , 2016, Scientific Reports.

[126]  P. Thompson,et al.  Nicotine induces neutrophil extracellular traps , 2016, Journal of leukocyte biology.

[127]  Mengsen Li,et al.  Residues Responsible for the Selectivity of α-Conotoxins for Ac-AChBP or nAChRs , 2016, Marine drugs.

[128]  C. Ghelardini,et al.  The α9α10 nicotinic receptor antagonist α-conotoxin RgIA prevents neuropathic pain induced by oxaliplatin treatment , 2016, Experimental Neurology.

[129]  V. N. Mal'tseva,et al.  Nicotinic receptor involvement in regulation of functions of mouse neutrophils from inflammatory site. , 2016, Immunobiology.

[130]  W. Padberg,et al.  Phosphocholine – an agonist of metabotropic but not of ionotropic functions of α9-containing nicotinic acetylcholine receptors , 2016, Scientific Reports.

[131]  C. Bouzat,et al.  Expression and Functional Role of α7 Nicotinic Receptor in Human Cytokine-stimulated Natural Killer (NK) Cells* , 2016, The Journal of Biological Chemistry.

[132]  D. Craik,et al.  Structure-Activity Studies of Cysteine-Rich α-Conotoxins that Inhibit High-Voltage-Activated Calcium Channels via GABA(B) Receptor Activation Reveal a Minimal Functional Motif. , 2016, Angewandte Chemie.

[133]  Dongting Zhangsun,et al.  Anti-hypersensitive effect of intramuscular administration of αO-conotoxin GeXIVA[1,2] and GeXIVA[1,4] in rats of neuropathic pain , 2016, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[134]  J. Hao,et al.  Infiltration of CCR2+Ly6Chigh Proinflammatory Monocytes and Neutrophils into the Central Nervous System Is Modulated by Nicotinic Acetylcholine Receptors in a Model of Multiple Sclerosis , 2016, The Journal of Immunology.

[135]  J. Hao,et al.  Nicotinic Acetylcholine Receptors Modulate Bone Marrow-Derived Pro-Inflammatory Monocyte Production and Survival , 2016, PloS one.

[136]  D. Dolgikh,et al.  Human Secreted Ly-6/uPAR Related Protein-1 (SLURP-1) Is a Selective Allosteric Antagonist of α7 Nicotinic Acetylcholine Receptor , 2016, PloS one.

[137]  R. Coimbra,et al.  Up-regulation of the human-specific CHRFAM7A gene in inflammatory bowel disease , 2016, BBA clinical.

[138]  J. Lasalde-Dominicci,et al.  The α7-nicotinic receptor is upregulated in immune cells from HIV-seropositive women: consequences to the cholinergic anti-inflammatory response , 2015, Clinical & translational immunology.

[139]  K. Kawashima,et al.  Non-neuronal cholinergic system in regulation of immune function with a focus on α7 nAChRs. , 2015, International immunopharmacology.

[140]  M. Christie,et al.  Conotoxin Interactions with α9α10-nAChRs: Is the α9α10-Nicotinic Acetylcholine Receptor an Important Therapeutic Target for Pain Management? , 2015, Toxins.

[141]  W. Padberg,et al.  Phosphocholine-Modified Macromolecules and Canonical Nicotinic Agonists Inhibit ATP-Induced IL-1β Release , 2015, The Journal of Immunology.

[142]  J. McIntosh,et al.  αS-conotoxin GVIIIB potently and selectively blocks α9α10 nicotinic acetylcholine receptors. , 2015, Biochemical pharmacology.

[143]  D. Craik,et al.  Cloning, synthesis, and characterization of αO-conotoxin GeXIVA, a potent α9α10 nicotinic acetylcholine receptor antagonist , 2015, Proceedings of the National Academy of Sciences.

[144]  W. Zang,et al.  Acetylcholine Inhibits LPS-Induced MMP-9 Production and Cell Migration via the a7 nAChR-JAK2/STAT3 Pathway in RAW264.7 Cells , 2015, Cellular Physiology and Biochemistry.

[145]  Qiuyun Dai,et al.  Structural and Functional Characterization of a Novel α-Conotoxin Mr1.7 from Conus marmoreus Targeting Neuronal nAChR α3β2, α9α10 and α6/α3β2β3 Subtypes , 2015, Marine drugs.

[146]  V. Tsetlin Three-finger snake neurotoxins and Ly6 proteins targeting nicotinic acetylcholine receptors: pharmacological tools and endogenous modulators. , 2015, Trends in pharmacological sciences.

[147]  M. Nouri-Shirazi,et al.  Nicotine Exposure Alters the mRNA Expression of Notch Ligands in Dendritic Cells and Their Response to Th1‐/Th2‐Promoting Stimuli , 2015, Scandinavian journal of immunology.

[148]  David John Adams,et al.  Novel Mechanism of Voltage-Gated N-type (Cav2.2) Calcium Channel Inhibition Revealed through α-Conotoxin Vc1.1 Activation of the GABAB Receptor , 2015, Molecular Pharmacology.

[149]  Andrew B. Wright,et al.  Limited Efficacy of α-Conopeptides, Vc1.1 and RgIA, To Inhibit Sensory Neuron CaV Current,, , 2015, eNeuro.

[150]  W. Kummer,et al.  Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion , 2015, Brain Structure and Function.

[151]  J. L. Le Caer,et al.  Isolation, purification and functional characterization of alpha-BnIA from Conus bandanus venom. , 2014, Toxicon : official journal of the International Society on Toxinology.

[152]  P. Herdewijn,et al.  Discovery of a new subclass of α-conotoxins in the venom of Conus australis. , 2014, Toxicon : official journal of the International Society on Toxinology.

[153]  M. J. McIntosh,et al.  α-Conotoxin RgIA protects against the development of nerve injury-induced chronic pain and prevents both neuronal and glial derangement , 2014, PAIN®.

[154]  H. Lester,et al.  The Duplicated α7 Subunits Assemble and Form Functional Nicotinic Receptors with the Full-length α7* , 2014, The Journal of Biological Chemistry.

[155]  S. Grando,et al.  Anti-Inflammatory Effects of the Nicotinergic Peptides SLURP-1 and SLURP-2 on Human Intestinal Epithelial Cells and Immunocytes , 2014, BioMed research international.

[156]  J. Tytgat,et al.  Conotoxins Targeting Nicotinic Acetylcholine Receptors: An Overview , 2014, Marine drugs.

[157]  D. Craik,et al.  Discovery, synthesis, and structure-activity relationships of conotoxins. , 2014, Chemical reviews.

[158]  H. Sunaga,et al.  SLURP-1, an endogenous α7 nicotinic acetylcholine receptor allosteric ligand, is expressed in CD205+ dendritic cells in human tonsils and potentiates lymphocytic cholinergic activity , 2014, Journal of Neuroimmunology.

[159]  I. Vetter,et al.  MrIC, a novel α-conotoxin agonist in the presence of PNU at endogenous α7 nicotinic acetylcholine receptors. , 2014, Biochemistry.

[160]  M. Christie,et al.  α9-nicotinic acetylcholine receptors contribute to the maintenance of chronic mechanical hyperalgesia, but not thermal or mechanical allodynia , 2014, Molecular pain.

[161]  P. Herdewijn,et al.  Structure-Function Elucidation of a New α-Conotoxin, Lo1a, from Conus longurionis , 2013, The Journal of Biological Chemistry.

[162]  D. Craik,et al.  Isolation and characterization of α-conotoxin LsIA with potent activity at nicotinic acetylcholine receptors. , 2013, Biochemical pharmacology.

[163]  R. Norton,et al.  Dicarba α-conotoxin Vc1.1 analogues with differential selectivity for nicotinic acetylcholine and GABAB receptors. , 2013, ACS chemical biology.

[164]  D. Craik,et al.  Determination of the α-conotoxin Vc1.1 binding site on the α9α10 nicotinic acetylcholine receptor. , 2013, Journal of medicinal chemistry.

[165]  F. Shi,et al.  Nicotinic Receptor β2 Determines NK Cell-Dependent Metastasis in a Murine Model of Metastatic Lung Cancer , 2013, PloS one.

[166]  T. Mak,et al.  Lymphocyte-derived ACh regulates local innate but not adaptive immunity , 2013, Proceedings of the National Academy of Sciences.

[167]  L. Azam,et al.  Molecular basis for the differential sensitivity of rat and human α9α10 nAChRs to α‐conotoxin RgIA , 2012, Journal of neurochemistry.

[168]  David John Adams,et al.  Intrathecal α-conotoxins Vc1.1, AuIB and MII acting on distinct nicotinic receptor subtypes reverse signs of neuropathic pain , 2012, Neuropharmacology.

[169]  David John Adams,et al.  γ-Aminobutyric Acid Type B (GABAB) Receptor Expression Is Needed for Inhibition of N-type (Cav2.2) Calcium Channels by Analgesic α-Conotoxins* , 2012, The Journal of Biological Chemistry.

[170]  David John Adams,et al.  Analgesic conotoxins: block and G protein‐coupled receptor modulation of N‐type (CaV2.2) calcium channels , 2012, British journal of pharmacology.

[171]  E. P. van der Zanden,et al.  Nicotinic Acetylcholine Receptor Expression and Susceptibility to Cholinergic Immunomodulation in Human Monocytes of Smoking Individuals , 2012, Neuroimmunomodulation.

[172]  D. Craik,et al.  RegIIA: an α4/7-conotoxin from the venom of Conus regius that potently blocks α3β4 nAChRs. , 2012, Biochemical pharmacology.

[173]  D. Palange,et al.  β2‐Adrenoreceptors of regulatory lymphocytes are essential for vagal neuromodulation of the innate immune system , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[174]  L. Dwoskin,et al.  The novel small molecule α9α10 nicotinic acetylcholine receptor antagonist ZZ-204G is analgesic. , 2011, European journal of pharmacology.

[175]  D. Bertrand,et al.  The chimeric gene CHRFAM7A, a partial duplication of the CHRNA7 gene, is a dominant negative regulator of α7*nAChR function. , 2011, Biochemical pharmacology.

[176]  Tak W. Mak,et al.  Acetylcholine-Synthesizing T Cells Relay Neural Signals in a Vagus Nerve Circuit , 2011, Science.

[177]  V. Tsetlin,et al.  Design of New α-Conotoxins: From Computer Modeling to Synthesis of Potent Cholinergic Compounds , 2011, Marine drugs.

[178]  H. Kurzen,et al.  The cholinergic system in guttate psoriasis with special reference to mast cells , 2011, Experimental dermatology.

[179]  R. Norton,et al.  Embryonic Toxin Expression in the Cone Snail Conus victoriae , 2011, The Journal of Biological Chemistry.

[180]  E. Deitch,et al.  α7-Cholinergic Receptor Mediates Vagal Induction of Splenic Norepinephrine , 2011, The Journal of Immunology.

[181]  V. Tsetlin,et al.  Assembly of nicotinic and other Cys‐loop receptors , 2011, Journal of neurochemistry.

[182]  D. Craik,et al.  Structure and Activity of α-Conotoxin PeIA at Nicotinic Acetylcholine Receptor Subtypes and GABAB Receptor-coupled N-type Calcium Channels* , 2011, The Journal of Biological Chemistry.

[183]  D. Bertrand,et al.  NMR Structure and Action on Nicotinic Acetylcholine Receptors of Water-soluble Domain of Human LYNX1* , 2011, The Journal of Biological Chemistry.

[184]  J. Renart,et al.  Function of partially duplicated human α77 nicotinic receptor subunit CHRFAM7A gene: potential implications for the cholinergic anti-inflammatory response. , 2011, The Journal of biological chemistry.

[185]  Yong-ming Yao,et al.  Stimulation of α7 Nicotinic Acetylcholine Receptor by Nicotine Increases Suppressive Capacity of Naturally Occurring CD4+CD25+ Regulatory T Cells in Mice In Vitro , 2010, Journal of Pharmacology and Experimental Therapeutics.

[186]  P. Whiteaker,et al.  Alexa Fluor 546‐ArIB[V11L;V16A] is a potent ligand for selectively labeling α7 nicotinic acetylcholine receptors , 2010, Journal of neurochemistry.

[187]  R. Langley,et al.  Nicotine Inhibits FcεRI-Induced Cysteinyl Leukotrienes and Cytokine Production without Affecting Mast Cell Degranulation Through α7/α9/α10-Nicotinic Receptors , 2010, The Journal of Immunology.

[188]  S. Grando,et al.  Auto/paracrine control of inflammatory cytokines by acetylcholine in macrophage-like U937 cells through nicotinic receptors. , 2010, International immunopharmacology.

[189]  S. Baksh,et al.  Nicotine-mediated signals modulate cell death and survival of T lymphocytes. , 2010, Toxicology and applied pharmacology.

[190]  David John Adams,et al.  Analgesic α-conotoxins Vc1.1 and Rg1A inhibit N-type calcium channels in sensory neurons of α9 nicotinic receptor knockout mice , 2010, Channels.

[191]  N. Parameswaran,et al.  Tumor necrosis factor-α signaling in macrophages. , 2010, Critical reviews in eukaryotic gene expression.

[192]  A. Malik,et al.  Requisite Role of the Cholinergic α7 Nicotinic Acetylcholine Receptor Pathway in Suppressing Gram-Negative Sepsis-Induced Acute Lung Inflammatory Injury , 2009, The Journal of Immunology.

[193]  A. Orr-Urtreger,et al.  Activation of the Cholinergic Anti-Inflammatory System by Nicotine Attenuates Neuroinflammation via Suppression of Th1 and Th17 Responses , 2009, The Journal of Immunology.

[194]  Richard A Houghten,et al.  A Synthetic Combinatorial Strategy for Developing α-Conotoxin Analogs as Potent α7 Nicotinic Acetylcholine Receptor Antagonists* , 2009, The Journal of Biological Chemistry.

[195]  N. Absalom,et al.  Alpha9 nicotinic acetylcholine receptors and the treatment of pain. , 2009, Biochemical pharmacology.

[196]  Natsuko Kageyama-Yahara,et al.  IgE-induced degranulation of mucosal mast cells is negatively regulated via nicotinic acetylcholine receptors. , 2008, Biochemical and biophysical research communications.

[197]  Alison Haythornthwaite,et al.  Analgesic α-Conotoxins Vc1.1 and Rg1A Inhibit N-Type Calcium Channels in Rat Sensory Neurons via GABAB Receptor Activation , 2008, The Journal of Neuroscience.

[198]  Kevin J. Tracey,et al.  Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia , 2008, Proceedings of the National Academy of Sciences.

[199]  S. Wonnacott,et al.  αConotoxin Arenatus IB[V11L,V16D] Is a Potent and Selective Antagonist at Rat and Human Native α7 Nicotinic Acetylcholine Receptors , 2008, Journal of Pharmacology and Experimental Therapeutics.

[200]  Eric Vivier,et al.  Functions of natural killer cells , 2008, Nature Immunology.

[201]  A. Collins,et al.  Synthesis and Characterization of 125I-α-Conotoxin ArIB[V11L;V16A], a Selective α7 Nicotinic Acetylcholine Receptor Antagonist , 2008, Journal of Pharmacology and Experimental Therapeutics.

[202]  D. Craik,et al.  Are α9α10 Nicotinic Acetylcholine Receptors a Pain Target for α-Conotoxins? , 2007, Molecular Pharmacology.

[203]  K. Kawashima,et al.  Enhanced serum antigen-specific IgG1 and proinflammatory cytokine production in nicotinic acetylcholine receptor α7 subunit gene knockout mice , 2007, Journal of Neuroimmunology.

[204]  H. Kurzen,et al.  Nicotinic receptors on rat alveolar macrophages dampen ATP-induced increase in cytosolic calcium concentration , 2007, Autonomic Neuroscience.

[205]  J. McIntosh,et al.  Targeting the α9α10 nicotinic acetylcholine receptor to treat severe pain , 2007 .

[206]  T. van der Poll,et al.  LOCAL STIMULATION OF α7 CHOLINERGIC RECEPTORS INHIBITS LPS-INDUCED TNF-α RELEASE IN THE MOUSE LUNG , 2007, Shock.

[207]  J. Changeux,et al.  The role of nicotinic receptors in B-lymphocyte development and activation. , 2007, Life sciences.

[208]  D. Yoshikami,et al.  Discovery, synthesis, and structure activity of a highly selective alpha7 nicotinic acetylcholine receptor antagonist. , 2007, Biochemistry.

[209]  M. Matthay,et al.  Activation of the α7 nAChR Reduces Acid-Induced Acute Lung Injury in Mice and Rats , 2007 .

[210]  M. Dolan,et al.  Molecular mechanisms of resistance and toxicity associated with platinating agents. , 2007, Cancer treatment reviews.

[211]  P. Williams,et al.  Nicotinic acetylcholine receptors on basophils and mast cells , 2006, Anaesthesia.

[212]  T. Yoshino,et al.  Effect of nicotine on IL‐18‐initiated immune response in human monocytes , 2006, Journal of leukocyte biology.

[213]  J. McIntosh,et al.  Molecular mechanism for analgesia involving specific antagonism of α9α10 nicotinic acetylcholine receptors , 2006, Proceedings of the National Academy of Sciences.

[214]  P. Sansonetti The innate signaling of dangers and the dangers of innate signaling , 2006, Nature Immunology.

[215]  Igor Tsigelny,et al.  α-Conotoxin OmIA Is a Potent Ligand for the Acetylcholine-binding Protein as Well as α3β2 and α7 Nicotinic Acetylcholine Receptors* , 2006, Journal of Biological Chemistry.

[216]  D. Craik,et al.  The Synthesis, Structural Characterization, and Receptor Specificity of the α-Conotoxin Vc1.1* , 2006, Journal of Biological Chemistry.

[217]  D. Tracey,et al.  Immune and inflammatory mechanisms in neuropathic pain , 2006, Brain Research Reviews.

[218]  V. Pavlov,et al.  Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis , 2006, The Journal of experimental medicine.

[219]  J. McIntosh,et al.  α-RgIA: A Novel Conotoxin That Specifically and Potently Blocks the α9α10 nAChR†,‡ , 2006 .

[220]  J. Changeux,et al.  The role of nicotinic acetylcholine receptors in lymphocyte development , 2006, Journal of Neuroimmunology.

[221]  T. Yoshino,et al.  alpha7 Nicotinic acetylcholine receptor stimulation inhibits lipopolysaccharide-induced interleukin-18 and -12 production in monocytes. , 2006, Journal of pharmacological sciences.

[222]  K. Gayler,et al.  Alpha-conotoxin Vc1.1 alleviates neuropathic pain and accelerates functional recovery of injured neurones , 2005, Brain Research.

[223]  C. Dejong,et al.  Nutritional stimulation of cholecystokinin receptors inhibits inflammation via the vagus nerve , 2005, The Journal of experimental medicine.

[224]  S. Kracun,et al.  Effects of Quinine, Quinidine, and Chloroquine on α9α10 Nicotinic Cholinergic Receptors , 2005, Molecular Pharmacology.

[225]  J. McIntosh,et al.  A Novel α-Conotoxin, PeIA, Cloned from Conus pergrandis, Discriminates between Rat α9α10 and α7 Nicotinic Cholinergic Receptors* , 2005, Journal of Biological Chemistry.

[226]  Jian-Lan Jiang,et al.  [Effect of acetylcholine on the cytotoxicity of natural killer cells]. , 2005, Zhongguo ying yong sheng li xue za zhi = Zhongguo yingyong shenglixue zazhi = Chinese journal of applied physiology.

[227]  H. Berthoud,et al.  Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway , 2005, Nature Immunology.

[228]  J. Changeux,et al.  Nicotinic receptors regulate B lymphocyte activation and immune response. , 2005, European journal of pharmacology.

[229]  N. Unwin,et al.  Refined structure of the nicotinic acetylcholine receptor at 4A resolution. , 2005, Journal of molecular biology.

[230]  M. Rustici,et al.  Sequence homology between HIV gp120, rabies virus glycoprotein, and snake venom neurotoxins , 2005, Archives of Virology.

[231]  S. Sine,et al.  α-Conotoxins ImI and ImII target distinct regions of the human α7 nicotinic acetylcholine receptor and distinguish human nicotinic receptor subtypes , 2004 .

[232]  R. Ferris,et al.  Characterization of the human nicotinic acetylcholine receptor subunit alpha (alpha) 9 (CHRNA9) and alpha (alpha) 10 (CHRNA10) in lymphocytes. , 2004, Life sciences.

[233]  David John Adams,et al.  Chemical and functional identification and characterization of novel sulfated alpha-conotoxins from the cone snail Conus anemone. , 2004, Journal of medicinal chemistry.

[234]  Laure Plantard,et al.  Identification of SLURP-1 as an epidermal neuromodulator explains the clinical phenotype of Mal de Meleda. , 2003, Human molecular genetics.

[235]  A. Maelicke,et al.  α‐Conotoxins EpI and AuIB switch subtype selectivity and activity in native versus recombinant nicotinic acetylcholine receptors , 2003 .

[236]  M. Nouri-Shirazi,et al.  Evidence for the immunosuppressive role of nicotine on human dendritic cell functions , 2003, Immunology.

[237]  N. Hogg Nicotine has suppressive effects on dendritic cell function , 2003, Immunology.

[238]  P. Gopalakrishnakone,et al.  Non-conventional toxins from Elapid venoms. , 2003, Toxicon : official journal of the International Society on Toxinology.

[239]  D. Craik,et al.  Isolation, Structure, and Activity of GID, a Novel α4/7-Conotoxin with an Extended N-terminal Sequence* , 2003, The Journal of Biological Chemistry.

[240]  J. McIntosh,et al.  α-Conotoxins ImI and ImII , 2003, The Journal of Biological Chemistry.

[241]  Kevin J. Tracey,et al.  Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation , 2002, Nature.

[242]  Kevin J. Tracey,et al.  The inflammatory reflex , 2002, Nature.

[243]  W. Kummer,et al.  Coexpression of α9 and α10 nicotinic acetylcholine receptors in rat dorsal root ganglion neurons , 2002, Neuroscience.

[244]  T. Sixma,et al.  Crystal structure of an ACh-binding protein reveals the ligand-binding domain of nicotinic receptors , 2001, Nature.

[245]  D Bertrand,et al.  “Weak Toxin” from Naja kaouthia Is a Nontoxic Antagonist of α7 and Muscle-type Nicotinic Acetylcholine Receptors* , 2001, The Journal of Biological Chemistry.

[246]  K. Tracey,et al.  Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin , 2000, Nature.

[247]  S. Wattler,et al.  Structural and phylogenetic characterization of human SLURP‐1, the first secreted mammalian member of the Ly‐6/uPAR protein superfamily , 2008, Protein science : a publication of the Protein Society.

[248]  D. Yoshikami,et al.  Single-Residue Alteration in α-Conotoxin PnIA Switches Its nAChR Subtype Selectivity† , 1999 .

[249]  A. Sali,et al.  lynx1, an Endogenous Toxin-like Modulator of Nicotinic Acetylcholine Receptors in the Mammalian CNS , 1999, Neuron.

[250]  P. Alewood,et al.  α‐Conotoxin ImI Inhibits the α‐Bungarotoxin‐Resistant Nicotinic Response in Bovine Adrenal Chromaffin Cells , 1999 .

[251]  G. Cragg Paclitaxel (Taxol®): A success story with valuable lessons for natural product drug discovery and development , 1998, Medicinal research reviews.

[252]  K. Kawashima,et al.  Induction of choline acetyltransferase mRNA in human mononuclear leukocytes stimulated by phytohemagglutinin, a T-cell activator , 1998, Journal of Neuroimmunology.

[253]  N. Unwin The Nicotinic Acetylcholine Receptor of theTorpedoElectric Ray , 1998 .

[254]  K. Kawashima,et al.  Localization and synthesis of acetylcholine in human leukemic T cell lines , 1996, Journal of neuroscience research.

[255]  M. Rustici,et al.  Binding of HIV‐1 gp120 to the nicotinic receptor , 1992, FEBS letters.

[256]  E. Masini,et al.  Release of histamine from rat mast cells by acetylcholine , 1978, Nature.

[257]  Y. Dupont X‐ray diffraction study of membrane fragments rich in acetylcholine receptor protein prepared from the electric organ of Torpedo Marmorata , 1974, FEBS letters.