1
|
Parodi J, Mira RG, Martinez-Torres A, Inestrosa NC. Wnt-5a/Ca 2+ pathway modulates endogenous current and oocyte structure of Xenopus laevis. Biochem Biophys Res Commun 2024; 738:150507. [PMID: 39154550 DOI: 10.1016/j.bbrc.2024.150507] [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: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Wnt signaling plays an essential role in cellular processes like development, maturation, and function maintenance. Xenopus laevis oocytes are a suitable model to study not only the development but also the function of different receptors expressed in their membranes, like those receptors expressed in the central nervous system (CNS) including Frizzled 7. Here, using frog oocytes and recordings of endogenous membrane currents in a two-electrode path configuration along with morphological observations, we evaluated the role of the non-canonical Wnt-5a ligand in oocytes. We found that acute application of Wnt-5a generated changes in endogenous calcium-dependent currents, entry oscillatory current, the membrane's outward current, and induced membrane depolarization. The incubation of oocytes with Wnt-5a caused a reduction of the membrane potential, potassium outward current, and protected the ATP current in the epithelium/theca removed (ETR) model. The oocytes exposed to Wnt-5a showed increased viability and an increase in the percentage of the germinal vesicle breakdown (GVBD), at a higher level than the control with progesterone. Altogether, our results suggest that Wnt-5a modulates different aspects of oocyte structure and generates calcium-dependent endogenous current alteration and GVDB process with a change in membrane potential at different concentrations and times of the exposition. These results help to understand the cellular effect of Wnt-5a and present the use of Xenopus oocytes to explore the mechanism that could impact the activation of Wnt signaling.
Collapse
Affiliation(s)
- Jorge Parodi
- Departmento de Análisis de Datos, Facultad de Ciencias Sociales, Universidad Autónoma de Chile, Temuco, Chile; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Ataulfo Martinez-Torres
- Laboratorio de Neurobiología Molecular y Celular, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla-Querétaro, UNAM, Mexico
| | - Nibaldo C Inestrosa
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
2
|
Ghosh A, Ribeiro-Rodrigues L, Ruffolo G, Alfano V, Domingos C, Rei N, Tosh DK, Rombo DM, Morais TP, Valente CA, Xapelli S, Bordadágua B, Rainha-Campos A, Bentes C, Aronica E, Diógenes MJ, Vaz SH, Ribeiro JA, Palma E, Jacobson KA, Sebastião AM. Selective modulation of epileptic tissue by an adenosine A 3 receptor-activating drug. Br J Pharmacol 2024; 181:5041-5061. [PMID: 39300608 DOI: 10.1111/bph.17319] [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: 03/27/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Adenosine, through the A1 receptor (A1R), is an endogenous anticonvulsant. The development of adenosine receptor agonists as antiseizure medications has been hampered by their cardiac side effects. A moderately A1R-selective agonist, MRS5474, has been reported to suppress seizures without considerable cardiac action. Hypothesizing that this drug could act through other than A1R and/or through a disease-specific mechanism, we assessed the effect of MRS5474 on the hippocampus. EXPERIMENTAL APPROACH Excitatory synaptic currents, field potentials, spontaneous activity, [3H]GABA uptake and GABAergic currents were recorded from rodent or human hippocampal tissue. Alterations in adenosine A3 receptor (A3R) density in human tissue were assessed by Western blot. KEY RESULTS MRS5474 (50-500 nM) was devoid of effect upon rodent excitatory synaptic signals in hippocampal slices, except when hyperexcitability was previously induced in vivo or ex vivo. MRS5474 inhibited GABA transporter type 1 (GAT-1)-mediated γ-aminobutyric acid (GABA) uptake, an action not blocked by an A1R antagonist but blocked by an A3R antagonist and mimicked by an A3R agonist. A3R was overexpressed in human hippocampal tissue samples from patients with epilepsy that had focal resection from surgery. MRS5474 induced a concentration-dependent potentiation of GABA-evoked currents in oocytes micro-transplanted with human hippocampal membranes prepared from epileptic hippocampal tissue but not from non-epileptic tissue, an action blocked by an A3R antagonist. CONCLUSION AND IMPLICATIONS We identified a drug that activates A3R and has selective actions on epileptic hippocampal tissue. This underscores A3R as a promising target for the development of antiseizure medications.
Collapse
Affiliation(s)
- Anwesha Ghosh
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS San Raffaele Roma, Rome, Italy
| | | | - Cátia Domingos
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Tatiana P Morais
- Neuroscience Division, School of Bioscience, Cardiff University, Cardiff, UK
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Beatriz Bordadágua
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
- Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Alexandre Rainha-Campos
- Centro de Referência para a área da Epilepsia Refratária (ERN EpiCARE Member), CHULN, Lisbon, Portugal
- Serviço de Neurologia, CHULN, Lisbon, Portugal
| | - Carla Bentes
- Centro de Referência para a área da Epilepsia Refratária (ERN EpiCARE Member), CHULN, Lisbon, Portugal
- Laboratório de EEG/Sono-Unidade de Monitorização Neurofisiológica, Serviço de Neurologia, CHULN, Lisbon, Portugal
- Centro de Estudos Egas Moniz, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Eleonora Aronica
- Amsterdam Neuroscience, Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
3
|
Trobec T, Lamassiaude N, Benoit E, Žužek MC, Sepčić K, Kladnik J, Turel I, Aráoz R, Frangež R. New insights into the effects of organometallic ruthenium complexes on nicotinic acetylcholine receptors. Chem Biol Interact 2024; 402:111213. [PMID: 39209017 DOI: 10.1016/j.cbi.2024.111213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are expressed in excitable and non-excitable cells of the organism. Extensive studies suggest that nAChR ligands have therapeutic potential, notably for neurological and psychiatric disorders. Organometallic ruthenium complexes are known to inhibit several medically important enzymes such as cholinesterases. In addition, they can also interact with muscle- and neuronal-subtype nAChRs. The present study aimed to investigate the direct effects of three organometallic ruthenium complexes, [(η6-p-cymene)Ru(II)(5-nitro-1,10-phenanthroline)Cl]Cl (C1-Cl), [(η6-p-cymene)Ru(II)(1-hydroxypyridine-2(1H)-thionato)Cl] (C1a) and [(η6-p-cymene)Ru(II)(1-hydroxy-3-methoxypyridine-2(1H)-thionato)pta]PF6 (C1), on muscle-subtype (Torpedo) nAChRs and on the two most abundant human neuronal-subtype nAChRs in the CNS (α4β2 and α7) expressed in Xenopus laevis oocytes, using the two-electrode voltage-clamp. The results show that none of the three compounds had agonistic activity on any of the nAChR subtypes studied. In contrast, C1-Cl reversibly blocked Torpedo nAChR (half-reduction of ACh-evoked peak current amplitude by 332 nM of compound). When tested at 10 μM, C1-Cl was statistically more potent to inhibit TorpedonAChR than α4β2 and α7 nAChRs. Similar results of C1 effects were obtained on Torpedo and α4β2 nAChRs, while no action of the compound was detected on α7 nAChRs. Finally, the effects of C1a were statistically similar on the three nAChR subtypes but, in contrast to C1-Cl and C1, the inhibition was hardly reversible. These results, together with our previous studies on isolated mouse neuromuscular preparations, strongly suggest that C1-Cl is, among the three compounds studied, the only molecule that could be used as a potential myorelaxant drug.
Collapse
Affiliation(s)
- Tomaž Trobec
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé (DMTS), Service d'Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, 91191 Gif-sur-Yvette, France; Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Nicolas Lamassiaude
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé (DMTS), Service d'Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, 91191 Gif-sur-Yvette, France
| | - Evelyne Benoit
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé (DMTS), Service d'Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, 91191 Gif-sur-Yvette, France
| | - Monika Cecilija Žužek
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Rómulo Aráoz
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé (DMTS), Service d'Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, 91191 Gif-sur-Yvette, France
| | - Robert Frangež
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
4
|
Bourne Y, Sulzenbacher G, Chabaud L, Aráoz R, Radić Z, Conrod S, Taylor P, Guillou C, Molgó J, Marchot P. The Cyclic Imine Core Common to the Marine Macrocyclic Toxins Is Sufficient to Dictate Nicotinic Acetylcholine Receptor Antagonism. Mar Drugs 2024; 22:149. [PMID: 38667766 PMCID: PMC11050823 DOI: 10.3390/md22040149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Macrocyclic imine phycotoxins are an emerging class of chemical compounds associated with harmful algal blooms and shellfish toxicity. Earlier binding and electrophysiology experiments on nAChR subtypes and their soluble AChBP surrogates evidenced common trends for substantial antagonism, binding affinities, and receptor-subtype selectivity. Earlier, complementary crystal structures of AChBP complexes showed that common determinants within the binding nest at each subunit interface confer high-affinity toxin binding, while distinctive determinants from the flexible loop C, and either capping the nest or extending toward peripheral subsites, dictate broad versus narrow receptor subtype selectivity. From these data, small spiroimine enantiomers mimicking the functional core motif of phycotoxins were chemically synthesized and characterized. Voltage-clamp analyses involving three nAChR subtypes revealed preserved antagonism for both enantiomers, despite lower subtype specificity and binding affinities associated with faster reversibility compared with their macrocyclic relatives. Binding and structural analyses involving two AChBPs pointed to modest affinities and positional variability of the spiroimines, along with a range of AChBP loop-C conformations denoting a prevalence of antagonistic properties. These data highlight the major contribution of the spiroimine core to binding within the nAChR nest and confirm the need for an extended interaction network as established by the macrocyclic toxins to define high affinities and marked subtype specificity. This study identifies a minimal set of functional pharmacophores and binding determinants as templates for designing new antagonists targeting disease-associated nAChR subtypes.
Collapse
Affiliation(s)
- Yves Bourne
- Lab “Architecture et Fonction des Macromolécules Biologiques” (AFMB), Aix-Marseille Univ, CNRS, Faculté des Sciences Campus Luminy, 13288 Marseille cedex 09, France; (Y.B.); (G.S.)
| | - Gerlind Sulzenbacher
- Lab “Architecture et Fonction des Macromolécules Biologiques” (AFMB), Aix-Marseille Univ, CNRS, Faculté des Sciences Campus Luminy, 13288 Marseille cedex 09, France; (Y.B.); (G.S.)
| | - Laurent Chabaud
- Institut de Chimie des Substances Naturelles (ICSN), Univ Paris-Saclay, CNRS, 91198 Gif-sur-Yvette, France; (L.C.); (C.G.)
| | - Rómulo Aráoz
- Service d’Ingénierie Moléculaire pour la Santé (SIMoS) EMR CNRS 9004, Département Médicaments et Technologies pour la Santé, Institut des Sciences du Vivant Frédéric Joliot, CEA, INRAE, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.A.); (J.M.)
| | - Zoran Radić
- Skaggs School of Pharmacy and Pharmaceutical Sciences (SSPPS), University of California San Diego, La Jolla, CA 92093-0751, USA; (Z.R.); (P.T.)
| | - Sandrine Conrod
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), Aix Marseille Univ, CNRS, 13344 Marseille, France;
| | - Palmer Taylor
- Skaggs School of Pharmacy and Pharmaceutical Sciences (SSPPS), University of California San Diego, La Jolla, CA 92093-0751, USA; (Z.R.); (P.T.)
| | - Catherine Guillou
- Institut de Chimie des Substances Naturelles (ICSN), Univ Paris-Saclay, CNRS, 91198 Gif-sur-Yvette, France; (L.C.); (C.G.)
| | - Jordi Molgó
- Service d’Ingénierie Moléculaire pour la Santé (SIMoS) EMR CNRS 9004, Département Médicaments et Technologies pour la Santé, Institut des Sciences du Vivant Frédéric Joliot, CEA, INRAE, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.A.); (J.M.)
| | - Pascale Marchot
- Lab “Architecture et Fonction des Macromolécules Biologiques” (AFMB), Aix-Marseille Univ, CNRS, Faculté des Sciences Campus Luminy, 13288 Marseille cedex 09, France; (Y.B.); (G.S.)
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), Aix Marseille Univ, CNRS, 13344 Marseille, France;
| |
Collapse
|
5
|
Ruffolo G, Gaeta A, Cannata B, Pinzaglia C, Aronica E, Morano A, Cifelli P, Palma E. GABAergic Neurotransmission in Human Tissues Is Modulated by Cannabidiol. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122042. [PMID: 36556407 PMCID: PMC9786817 DOI: 10.3390/life12122042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Recently, the potential use of phytocannabinoids (pCBs) to treat different pathological conditions has attracted great attention in the scientific community. Among the different pCBs, cannabidiol (CBD) has showed interesting biological properties, making it a promising molecule with a high security profile that has been approved for treatment as an add-on therapy in patients afflicted by severe pharmaco-resistant epilepsy, including Dravet syndrome (DS), Lennox-Gastaut syndrome (LGS) and tuberous sclerosis complex (TSC). CBD is pharmacologically considered a "dirty drug", since it has the capacity to bind different targets and to activate several cellular pathways. GABAergic impairment is one of the key processes during the epileptogenesis period able to induce a generalized hyperexcitability of the central nervous system (CNS), leading to epileptic seizures. Here, by using the microtransplantation of human brain membranes approach in Xenopus oocytes and electrophysiological recordings, we confirm the ability of CBD to modulate GABAergic neurotransmission in human cerebral tissues obtained from patients afflicted by different forms of pharmaco-resistant epilepsies, such as DS, TSC, focal cortical dysplasia (FCD) type IIb and temporal lobe epilepsy (TLE). Furthermore, using cDNAs encoding for human GABAA receptor subunits, we found that α1β2 receptors are still affected by CBD, while classical benzodiazepine lost its efficacy as expected.
Collapse
Affiliation(s)
- Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy
- IRCCS San Raffaele Roma, 00163 Rome, Italy
| | - Alessandro Gaeta
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy
| | - Beatrice Cannata
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy
| | - Camilla Pinzaglia
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland, 0397 Heemstede, The Netherlands
| | - Alessandra Morano
- Department of Human Neuroscience, University of Rome Sapienza, 00185 Rome, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Correspondence:
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy
- IRCCS San Raffaele Roma, 00163 Rome, Italy
| |
Collapse
|
6
|
Ruffolo G, Alfano V, Romagnolo A, Zimmer T, Mills JD, Cifelli P, Gaeta A, Morano A, Anink J, Mühlebner A, Vezzani A, Aronica E, Palma E. GABA A receptor function is enhanced by Interleukin-10 in human epileptogenic gangliogliomas and its effect is counteracted by Interleukin-1β. Sci Rep 2022; 12:17956. [PMID: 36289354 PMCID: PMC9605959 DOI: 10.1038/s41598-022-22806-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
Gangliogliomas (GGs) are low-grade brain tumours that cause intractable focal epilepsy in children and adults. In GG, as in epileptogenic focal malformations (i.e., tuberous sclerosis complex, TSC), there is evidence of sustained neuroinflammation with involvement of the pro-inflammatory cytokine IL-1β. On the other hand, anti-inflammatory mediators are less studied but bear relevance for understanding seizure mechanisms. Therefore, we investigated the effect of the key anti-inflammatory cytokine IL-10 on GABAergic neurotransmission in GG. We assessed the IL-10 dependent signaling by transcriptomic analysis, immunohistochemistry and performed voltage-clamp recordings on Xenopus oocytes microtransplanted with cell membranes from brain specimens, to overcome the limited availability of acute GG slices. We report that IL-10-related mRNAs were up-regulated in GG and slightly in TSC. Moreover, we found IL-10 receptors are expressed by neurons and astroglia. Furthermore, GABA currents were potentiated significantly by IL-10 in GG. This effect was time and dose-dependent and inhibited by blockade of IL-10 signaling. Notably, in the same tissue, IL-1β reduced GABA current amplitude and prevented the IL-10 effect. These results suggest that in epileptogenic tissue, pro-inflammatory mechanisms of hyperexcitability prevail over key anti-inflammatory pathways enhancing GABAergic inhibition. Hence, boosting the effects of specific anti-inflammatory molecules could resolve inflammation and reduce intractable seizures.
Collapse
Affiliation(s)
- Gabriele Ruffolo
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Veronica Alfano
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Alessia Romagnolo
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Till Zimmer
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - James D. Mills
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK ,grid.452379.e0000 0004 0386 7187Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Pierangelo Cifelli
- grid.158820.60000 0004 1757 2611Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Gaeta
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Alessandra Morano
- grid.7841.aDepartment of Human Neuroscience, University of Rome Sapienza, Rome, Italy
| | - Jasper Anink
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.7692.a0000000090126352Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annamaria Vezzani
- grid.4527.40000000106678902Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Eleonora Aronica
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.419298.f0000 0004 0631 9143Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Eleonora Palma
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| |
Collapse
|
7
|
Ivorra I, Alberola-Die A, Cobo R, González-Ros JM, Morales A. Xenopus Oocytes as a Powerful Cellular Model to Study Foreign Fully-Processed Membrane Proteins. MEMBRANES 2022; 12:986. [PMID: 36295745 PMCID: PMC9610954 DOI: 10.3390/membranes12100986] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
The use of Xenopus oocytes in electrophysiological and biophysical research constitutes a long and successful story, providing major advances to the knowledge of the function and modulation of membrane proteins, mostly receptors, ion channels, and transporters. Earlier reports showed that these cells are capable of correctly expressing heterologous proteins after injecting the corresponding mRNA or cDNA. More recently, the Xenopus oocyte has become an outstanding host-cell model to carry out detailed studies on the function of fully-processed foreign membrane proteins after their microtransplantation to the oocyte. This review focused on the latter overall process of transplanting foreign membrane proteins to the oocyte after injecting plasma membranes or purified and reconstituted proteins. This experimental approach allows for the study of both the function of mature proteins, with their native stoichiometry and post-translational modifications, and their putative modulation by surrounding lipids, mostly when the protein is purified and reconstituted in lipid matrices of defined composition. Remarkably, this methodology enables functional microtransplantation to the oocyte of membrane receptors, ion channels, and transporters from different sources including human post-mortem tissue banks. Despite the large progress achieved over the last decades on the structure, function, and modulation of neuroreceptors and ion channels in healthy and pathological tissues, many unanswered questions remain and, most likely, Xenopus oocytes will continue to help provide valuable responses.
Collapse
Affiliation(s)
- Isabel Ivorra
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Apdo 99, E-03080 Alicante, Spain
| | - Armando Alberola-Die
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Apdo 99, E-03080 Alicante, Spain
| | - Raúl Cobo
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Apdo 99, E-03080 Alicante, Spain
| | - José Manuel González-Ros
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, E-03202 Elche, Spain
| | - Andrés Morales
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Apdo 99, E-03080 Alicante, Spain
| |
Collapse
|
8
|
Bhatt M, Di Iacovo A, Romanazzi T, Roseti C, Cinquetti R, Bossi E. The "www" of Xenopus laevis Oocytes: The Why, When, What of Xenopus laevis Oocytes in Membrane Transporters Research. MEMBRANES 2022; 12:membranes12100927. [PMID: 36295686 PMCID: PMC9610376 DOI: 10.3390/membranes12100927] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 05/16/2023]
Abstract
After 50 years, the heterologous expression of proteins in Xenopus laevis oocytes is still essential in many research fields. New approaches and revised protocols, but also classical methods, such as the two-electrode voltage clamp, are applied in studying membrane transporters. New and old methods for investigating the activity and the expression of Solute Carriers (SLC) are reviewed, and the kinds of experiment that are still useful to perform with this kind of cell are reported. Xenopus laevis oocytes at the full-grown stage have a highly efficient biosynthetic apparatus that correctly targets functional proteins at the defined compartment. This small protein factory can produce, fold, and localize almost any kind of wild-type or recombinant protein; some tricks are required to obtain high expression and to verify the functionality. The methodologies examined here are mainly related to research in the field of membrane transporters. This work is certainly not exhaustive; it has been carried out to be helpful to researchers who want to quickly find suggestions and detailed indications when investigating the functionality and expression of the different members of the solute carrier families.
Collapse
Affiliation(s)
- Manan Bhatt
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Angela Di Iacovo
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Tiziana Romanazzi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Cristina Roseti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Centre for Neuroscience—Via Manara 7, University of Insubria, 21052 Busto Arsizio, Italy
| | - Raffaella Cinquetti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
| | - Elena Bossi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Centre for Neuroscience—Via Manara 7, University of Insubria, 21052 Busto Arsizio, Italy
- Correspondence:
| |
Collapse
|
9
|
Rousset M, Humez S, Laurent C, Buée L, Blum D, Cens T, Vignes M, Charnet P. Mammalian Brain Ca2+ Channel Activity Transplanted into Xenopus laevis Oocytes. MEMBRANES 2022; 12:membranes12050496. [PMID: 35629822 PMCID: PMC9146698 DOI: 10.3390/membranes12050496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023]
Abstract
Several mutations on neuronal voltage-gated Ca2+ channels (VGCC) have been shown to cause neurological disorders and contribute to the initiation of epileptic seizures, migraines, or cerebellar degeneration. Analysis of the functional consequences of these mutations mainly uses heterologously expressed mutated channels or transgenic mice which mimic these pathologies, since direct electrophysiological approaches on brain samples are not easily feasible. We demonstrate that mammalian voltage-gated Ca2+ channels from membrane preparation can be microtransplanted into Xenopus oocytes and can conserve their activity. This method, originally described to study the alteration of GABA receptors in human brain samples, allows the recording of the activity of membrane receptors and channels with their native post-translational processing, membrane environment, and regulatory subunits. The use of hippocampal, cerebellar, or cardiac membrane preparation displayed different efficacy for transplanted Ca2+ channel activity. This technique, now extended to the recording of Ca2+ channel activity, may therefore be useful in order to analyze the calcium signature of membrane preparations from unfixed human brain samples or normal and transgenic mice.
Collapse
Affiliation(s)
- Matthieu Rousset
- IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, 1919 Route de Mende, 34293 Montpellier, France; (T.C.); (M.V.)
- Correspondence: (M.R.); (P.C.); Tel.: +33-467-613-666 (M.R. & P.C.)
| | - Sandrine Humez
- Lille Neuroscience & Cognition, Université de Lille, F-59000 Lille, France; (S.H.); (C.L.); (L.B.); (D.B.)
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille, France
- Lille Neuroscience & Cognition, Alzheimer & Tauopathies, CHU-Lille, F-59000 Lille, France
| | - Cyril Laurent
- Lille Neuroscience & Cognition, Université de Lille, F-59000 Lille, France; (S.H.); (C.L.); (L.B.); (D.B.)
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille, France
- Lille Neuroscience & Cognition, Alzheimer & Tauopathies, CHU-Lille, F-59000 Lille, France
| | - Luc Buée
- Lille Neuroscience & Cognition, Université de Lille, F-59000 Lille, France; (S.H.); (C.L.); (L.B.); (D.B.)
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille, France
- Lille Neuroscience & Cognition, Alzheimer & Tauopathies, CHU-Lille, F-59000 Lille, France
| | - David Blum
- Lille Neuroscience & Cognition, Université de Lille, F-59000 Lille, France; (S.H.); (C.L.); (L.B.); (D.B.)
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille, France
- Lille Neuroscience & Cognition, Alzheimer & Tauopathies, CHU-Lille, F-59000 Lille, France
| | - Thierry Cens
- IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, 1919 Route de Mende, 34293 Montpellier, France; (T.C.); (M.V.)
| | - Michel Vignes
- IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, 1919 Route de Mende, 34293 Montpellier, France; (T.C.); (M.V.)
| | - Pierre Charnet
- IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, 1919 Route de Mende, 34293 Montpellier, France; (T.C.); (M.V.)
- Correspondence: (M.R.); (P.C.); Tel.: +33-467-613-666 (M.R. & P.C.)
| |
Collapse
|
10
|
Bregestovski P, Heuser J, Martínez-Torres A. Ricardo Miledi - Outstanding Neuroscientist of XX-XXI Centuries. Neuroscience 2020; 439:1-9. [PMID: 32620217 DOI: 10.1016/j.neuroscience.2020.04.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Piotr Bregestovski
- Institut de Neurosciences des Systemes, UMR INSERM 1106, Aix-Marseille Universite, Faculte de Medecine, 27 Bd Jean Moulin, 13005 Marseille, France
| | - John Heuser
- Section on Integrative Biophysics, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - Ataulfo Martínez-Torres
- Laboratory of Molecular & Cellular Neurobiology, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, Quurétaro, 76230, Mexico.
| |
Collapse
|
11
|
Witkin JM, Ping X, Cerne R, Mouser C, Jin X, Hobbs J, Tiruveedhula VVNPB, Li G, Jahan R, Rashid F, Kumar Golani L, Cook JM, Smith JL. The value of human epileptic tissue in the characterization and development of novel antiepileptic drugs: The example of CERC-611 and KRM-II-81. Brain Res 2019; 1722:146356. [PMID: 31369732 DOI: 10.1016/j.brainres.2019.146356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/17/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
The need for improved antiepileptics is clearly mandated despite the existence of multiple existing medicines from different chemical and mechanistic classes. Standard of care agents do not fully control epilepsies and have a variety of side-effect and safety issues. Patients typically take multiple antiepileptic drugs and yet many continue to have seizures. Antiepileptic-unresponsive seizures are life-disrupting and life-threatening. One approach to seizure control is surgical resection of affected brain tissue and associated neural circuits. Although non-human brain studies can provide insight into novel antiepileptic mechanisms, human epileptic brain is the bottom-line biological substrate. Human epileptic brain can provide definitive information on the presence or absence of the putative protein targets of interest in the patient population, the potential changes in these proteins in the epileptic state, and the engagement of novel molecules and their functional impact in target tissue. In this review, we discuss data on two novel potential antiepileptic drugs. CERC-611 (LY3130481) is an AMPA receptor antagonist that selectively blocks AMPA receptors associated with the auxiliary protein TARP γ-8 and is in clinical development. KRM-II-81 is a positive allosteric modulator of GABAA receptors selectively associated with protein subunits α2 and α 3. Preclinical data on these compounds argue that patient-based biological data increase the probability that a newly discovered molecule will translate its antiepileptic potential to patients.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| | - Xingjie Ping
- Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Rok Cerne
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Claire Mouser
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaoming Jin
- Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jon Hobbs
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Guanguan Li
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Rajwana Jahan
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Farjana Rashid
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Lalit Kumar Golani
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - James M Cook
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Ruffolo G, Cifelli P, Miranda-Lourenço C, De Felice E, Limatola C, Sebastião AM, Diógenes MJ, Aronica E, Palma E. Rare Diseases of Neurodevelopment: Maintain the Mystery or Use a Dazzling Tool for Investigation? The Case of Rett Syndrome. Neuroscience 2019; 439:146-152. [PMID: 31229630 DOI: 10.1016/j.neuroscience.2019.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 10/26/2022]
Abstract
The investigation on neurotransmission function during normal and pathologic development is a pivotal component needed to understand the basic mechanisms underlying neurodevelopmental pathologies. To study these diseases, many animal models have been generated which allowed to face the limited availability of human tissues and, as a consequence, most of the electrophysiology has been performed on these models of diseases. On the other hand, the technique of membrane microtransplantation in Xenopus oocytes allows the study of human functional neurotransmitter receptors thanks to the use of tissues from autopsies or surgeries, even in quantities that would not permit other kinds of functional studies. In this short article, we intend to underline how this technique is well-fit for the study of rare diseases by characterizing the electrophysiological properties of GABAA and AMPA receptors in Rett syndrome. For our purposes, we used both tissues from Rett syndrome patients and Mecp2-null mice, a well validated murine model of the same disease, in order to strengthen the solidity of our results through the comparison of the two. Our findings retrace previous results and, in the light of this, further argue in favor of Prof. Miledi's technique of membrane microtransplantation that proves itself a very useful tool of investigation in the field of neurophysiology. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
| | | | - Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Cristina Limatola
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy; IRCCS Neuromed, Pozzilli (IS), Italy
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), the Netherlands
| | - Eleonora Palma
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy.
| |
Collapse
|
13
|
Knopp KL, Simmons RMA, Guo W, Adams BL, Gardinier KM, Gernert DL, Ornstein PL, Porter W, Reel J, Ding C, Wang H, Qian Y, Burris KD, Need A, Barth V, Swanson S, Catlow J, Witkin JM, Zwart R, Sher E, Choong KC, Wall TM, Schober D, Felder CC, Kato AS, Bredt DS, Nisenbaum ES. Modulation of TARP γ8-Containing AMPA Receptors as a Novel Therapeutic Approach for Chronic Pain. J Pharmacol Exp Ther 2019; 369:345-363. [PMID: 30910921 DOI: 10.1124/jpet.118.250126] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/06/2019] [Indexed: 03/08/2025] Open
Abstract
Nonselective glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonists are efficacious in chronic pain but have significant tolerability issues, likely arising from the ubiquitous expression of AMPA receptors in the central nervous system (CNS). Recently, LY3130481 has been shown to selectively block AMPA receptors coassembled with the auxiliary protein, transmembrane AMPA receptor regulatory protein (TARP) γ8, which is highly expressed in the hippocampus but also in pain pathways, including anterior cingulate (ACC) and somatosensory cortices and the spinal cord, suggesting that selective blockade of γ8/AMPA receptors may suppress nociceptive signaling with fewer CNS side effects. The potency of LY3130481 on recombinant γ8-containing AMPA receptors was modulated by coexpression with other TARPs; γ2 subunits affected activity more than γ3 subunits. Consistent with these findings, LY3130481 had decreasing potency on receptors from rat hippocampal, cortical, spinal cord, and cerebellar neurons that was replicated in tissue from human brain. LY3130481 partially suppressed, whereas the nonselective AMPA antagonist GYKI53784 completely blocked, AMPA receptor-dependent excitatory postsynaptic potentials in ACC and spinal neurons in vitro. Similarly, LY3130481 attenuated short-term synaptic plasticity in spinal sensory neurons in vivo in response to stimulation of peripheral afferents. LY3130481 also significantly reduced nocifensive behaviors after intraplantar formalin that was correlated with occupancy of CNS γ8-containing AMPA receptors. In addition, LY3130481 dose-dependently attenuated established gait impairment after joint damage and tactile allodynia after spinal nerve ligation, all in the absence of motor side effects. Collectively, these data demonstrate that LY3130481 can suppress excitatory synaptic transmission and plasticity in pain pathways containing γ8/AMPA receptors and significantly reduce nocifensive behaviors, suggesting a novel, effective, and safer therapy for chronic pain conditions.
Collapse
Affiliation(s)
- Kelly L Knopp
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Rosa Maria A Simmons
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Wenhong Guo
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Benjamin L Adams
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Kevin M Gardinier
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Douglas L Gernert
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Paul L Ornstein
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Warren Porter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Jon Reel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Chunjin Ding
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - He Wang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Yuewei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Kevin D Burris
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Anne Need
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Vanessa Barth
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Steven Swanson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - John Catlow
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Ruud Zwart
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Emanuele Sher
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Kar-Chan Choong
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Theron M Wall
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Douglas Schober
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Christian C Felder
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Akihiko S Kato
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - David S Bredt
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| | - Eric S Nisenbaum
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (K.L.K., R.M.A.S., W.G., B.L.A., K.M.G., D.L.G., P.L.O., W.P., J.R., C.D., H.W., Y.Q., K.D.B., A.N., V.B., S.S., J.C., J.M.W., K.-C.C., T.M.W., D.S., C.C.F., A.S.K., D.S.B., E.S.N.) and Lilly Research Centre, Eli Lilly and Company Ltd., Erl Wood Manor, Windlesham, Surrey, United Kingdom (R.Z., E.S.)
| |
Collapse
|
14
|
Roseti C, Cifelli P, Ruffolo G, Barbieri E, Guescini M, Esposito V, Di Gennaro G, Limatola C, Giovannelli A, Aronica E, Palma E. Erythropoietin Increases GABA A Currents in Human Cortex from TLE Patients. Neuroscience 2019; 439:153-162. [PMID: 31047977 DOI: 10.1016/j.neuroscience.2019.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 11/15/2022]
Abstract
Erythropoietin (EPO) is a hematopoietic growth factor that has an important role in the erythropoiesis. EPO and its receptor (EPO-R) are expressed all over in the mammalian brain. Furthermore, it has been reported that EPO may exert neuroprotective effect in animal models of brain disorders as ischemia and epilepsy. Here, we investigate whether EPO could modulate the GABA-evoked currents (IGABA) in both human epileptic and non-epileptic control brain tissues. Therefore, we transplanted in Xenopus oocytes cell membranes obtained from autoptic and surgical brain tissues (cortex) of seven temporal lope epilepsy (TLE) patients and of five control patients. Two microelectrodes voltage-clamp technique has been used to record IGABA. Moreover, qRT-PCR assay was performed in the same human tissues to quantify the relative gene expression levels of EPO/EPO-R. To further confirm experiments in oocytes, we performed additional experiments using patch-clamp recording in slices obtained from rat cerebellum. We show that exposure to EPO significantly increased the amplitude of the IGABA in all the patients analyzed. No differences in the expression of EPO and EPO-R in both TLE and control patients have been found. Notably, the increase of IGABA has been recorded also in rat cerebellar slices. Our findings show a new modulatory action of EPO on GABAA receptors (GABAA-Rs). This effect could be relevant to balance the GABAergic dysfunction in human TLE. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
| | - Pierangelo Cifelli
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy
| | - Elena Barbieri
- Department of Biomolecular Science, University of Urbino Carlo Bo, Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Science, University of Urbino Carlo Bo, Urbino, Italy
| | | | | | - Cristina Limatola
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy; IRCCS Neuromed, Pozzilli, (IS), Italy
| | - Aldo Giovannelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila, Italy
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), the Netherlands
| | - Eleonora Palma
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, Rome, Italy.
| |
Collapse
|
15
|
A novel action of lacosamide on GABA A currents sets the ground for a synergic interaction with levetiracetam in treatment of epilepsy. Neurobiol Dis 2018; 115:59-68. [PMID: 29621596 DOI: 10.1016/j.nbd.2018.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/03/2018] [Accepted: 03/30/2018] [Indexed: 01/01/2023] Open
Abstract
Epilepsy is one of the most common chronic neurological diseases, and its pharmacological treatment holds great importance for both physicians and national authorities, especially considering the high proportion of drug-resistant patients (about 30%). Lacosamide (LCM) is an effective and well-tolerated new-generation antiepileptic drug (AED), currently licensed as add-on therapy for partial-onset seizures. However, LCM mechanism of action is still a matter of debate, although its effect on the voltage sensitive sodium channels is by far the most recognized. This study aimed to retrospectively analyze a cohort of 157 drug-resistant patients treated with LCM to describe the most common and effective therapeutic combinations and to investigate if the LCM can affect also GABAA-mediated neurotransmission as previously shown for levetiracetam (LEV). In our cohort, LEV resulted the compound most frequently associated with LCM in the responder subgroup. We therefore translated this clinical observation into the laboratory bench by taking advantage of the technique of "membrane micro-transplantation" in Xenopus oocytes and electrophysiological approaches to study human GABAA-evoked currents. In cortical brain tissues from refractory epileptic patients, we found that LCM reduces the use-dependent GABA impairment (i.e., "rundown") that it is considered one of the specific hallmarks of drug-resistant epilepsies. Notably, in line with our clinical observations, we found that the co-treatment with subthreshold concentrations of LCM and LEV, which had no effect on GABAA currents on their own, reduced GABA impairment in drug-resistant epileptic patients, and this effect was blocked by PKC inhibitors. Our findings demonstrate, for the first time, that LCM targets GABAA receptors and that it can act synergistically with LEV, improving the GABAergic function. This novel mechanism might contribute to explain the clinical efficacy of LCM-LEV combination in several refractory epileptic patients.
Collapse
|
16
|
Prorocentrolide-A from Cultured Prorocentrum lima Dinoflagellates Collected in Japan Blocks Sub-Types of Nicotinic Acetylcholine Receptors. Toxins (Basel) 2018; 10:toxins10030097. [PMID: 29495549 PMCID: PMC5869385 DOI: 10.3390/toxins10030097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 01/19/2023] Open
Abstract
Prorocentrolides are members of the cyclic imine phycotoxins family. Their chemical structure includes a 26-membered carbo-macrocycle and a 28-membered macrocyclic lactone arranged around a hexahydroisoquinoline that incorporates the characteristic cyclic imine group. Six prorocentrolides are already known. However, their mode of action remains undetermined. The aim of the present work was to explore whether prorocentrolide-A acts on nicotinic acetylcholine receptors (nAChRs), using competition-binding assays and electrophysiological techniques. Prorocentrolide-A displaced [125I]α-bungarotoxin binding to Torpedo membranes, expressing the muscle-type (α12β1γδ) nAChR, and in HEK-293 cells, expressing the chimeric chick neuronal α7-5HT3 nAChR. Functional studies revealed that prorocentrolide-A had no agonist action on nAChRs, but inhibited ACh-induced currents in Xenopus oocytes that had incorporated the muscle-type α12β1γδ nAChR to their membranes, or that expressed the human α7 nAChR, as revealed by voltage-clamp recordings. Molecular docking calculations showed the absence of the characteristic hydrogen bond between the iminium group of prorocentrolide-A and the backbone carbonyl group of Trp147 in the receptor, explaining its weaker affinity as compared to all other cyclic imine toxins. In conclusion, this is the first study to show that prorocentrolide-A acts on both muscle and neuronal nAChRs, but with higher affinity on the muscle-type nAChR.
Collapse
|
17
|
Ambrogini P, Albertini MC, Betti M, Galati C, Lattanzi D, Savelli D, Di Palma M, Saccomanno S, Bartolini D, Torquato P, Ruffolo G, Olivieri F, Galli F, Palma E, Minelli A, Cuppini R. Neurobiological Correlates of Alpha-Tocopherol Antiepileptogenic Effects and MicroRNA Expression Modulation in a Rat Model of Kainate-Induced Seizures. Mol Neurobiol 2018; 55:7822-7838. [PMID: 29468563 PMCID: PMC6132771 DOI: 10.1007/s12035-018-0946-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022]
Abstract
Seizure-triggered maladaptive neural plasticity and neuroinflammation occur during the latent period as a key underlying event in epilepsy chronicization. Previously, we showed that α-tocopherol (α-T) reduces hippocampal neuroglial activation and neurodegeneration in the rat model of kainic acid (KA)-induced status epilepticus (SE). These findings allowed us to postulate an antiepileptogenic potential for α-T in hippocampal excitotoxicity, in line with clinical evidence showing that α-T improves seizure control in drug-resistant patients. To explore neurobiological correlates of the α-T antiepileptogenic role, rats were injected with such vitamin during the latent period starting right after KA-induced SE, and the effects on circuitry excitability, neuroinflammation, neuronal death, and microRNA (miRNA) expression were investigated in the hippocampus. Results show that in α-T-treated epileptic rats, (1) the number of population spikes elicited by pyramidal neurons, as well as the latency to the onset of epileptiform-like network activity recover to control levels; (2) neuronal death is almost prevented; (3) down-regulation of claudin, a blood-brain barrier protein, is fully reversed; (4) neuroinflammation processes are quenched (as indicated by the decrease of TNF-α, IL-1β, GFAP, IBA-1, and increase of IL-6); (5) miR-146a, miR-124, and miR-126 expression is coherently modulated in hippocampus and serum by α-T. These findings support the potential of a timely intervention with α-T in clinical management of SE to reduce epileptogenesis, thus preventing chronic epilepsy development. In addition, we suggest that the analysis of miRNA levels in serum could provide clinicians with a tool to evaluate disease evolution and the efficacy of α-T therapy in SE.
Collapse
Affiliation(s)
- Patrizia Ambrogini
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy.
| | - Maria Cristina Albertini
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Michele Betti
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Claudia Galati
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Davide Lattanzi
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - David Savelli
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Michael Di Palma
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Stefania Saccomanno
- Department of Gastroenterology, Marche Polytechnic University, Ancona, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Fabiola Olivieri
- Department of Molecular and Clinical Sciences, Marche Polytechnic University, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS, Ancona, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Eleonora Palma
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Andrea Minelli
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, Section of Physiology, University of Urbino Carlo Bo, I-61029, Urbino, Italy
| |
Collapse
|
18
|
Shen W, Nan C, Nelson PT, Ripps H, Slaughter MM. GABA B receptor attenuation of GABA A currents in neurons of the mammalian central nervous system. Physiol Rep 2017; 5:5/6/e13129. [PMID: 28348006 PMCID: PMC5371550 DOI: 10.14814/phy2.13129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 11/18/2016] [Indexed: 11/24/2022] Open
Abstract
Ionotropic receptors are tightly regulated by second messenger systems and are often present along with their metabotropic counterparts on a neuron's plasma membrane. This leads to the hypothesis that the two receptor subtypes can interact, and indeed this has been observed in excitatory glutamate and inhibitory GABA receptors. In both systems the metabotropic pathway augments the ionotropic receptor response. However, we have found that the metabotropic GABAB receptor can suppress the ionotropic GABAA receptor current, in both the in vitro mouse retina and in human amygdala membrane fractions. Expression of amygdala membrane microdomains in Xenopus oocytes by microtransplantation produced functional ionotropic and metabotropic GABA receptors. Most GABAA receptors had properties of α‐subunit containing receptors, with ~5% having ρ‐subunit properties. Only GABAA receptors with α‐subunit‐like properties were regulated by GABAB receptors. In mouse retinal ganglion cells, where only α‐subunit‐containing GABAA receptors are expressed, GABAB receptors suppressed GABAA receptor currents. This suppression was blocked by GABAB receptor antagonists, G‐protein inhibitors, and GABAB receptor antibodies. Based on the kinetic differences between metabotropic and ionotropic receptors, their interaction would suppress repeated, rapid GABAergic inhibition.
Collapse
Affiliation(s)
- Wen Shen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine Florida Atlantic University, Boca Raton, Florida
| | - Changlong Nan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine Florida Atlantic University, Boca Raton, Florida
| | - Peter T Nelson
- Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky.,Sanders-Brown Centre on Aging, University of Kentucky, Lexington, Kentucky
| | - Harris Ripps
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois.,Whitman Investigator, Marine Biological Laboratory, Woods Hole, Massachusetts
| | - Malcolm M Slaughter
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
19
|
Molgó J, Marchot P, Aráoz R, Benoit E, Iorga BI, Zakarian A, Taylor P, Bourne Y, Servent D. Cyclic imine toxins from dinoflagellates: a growing family of potent antagonists of the nicotinic acetylcholine receptors. J Neurochem 2017; 142 Suppl 2:41-51. [PMID: 28326551 DOI: 10.1111/jnc.13995] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/24/2022]
Abstract
We present an overview of the toxicological profile of the fast-acting, lipophilic macrocyclic imine toxins, an emerging family of organic compounds associated with algal blooms, shellfish contamination and neurotoxicity. Worldwide, shellfish contamination incidents are expanding; therefore, the significance of these toxins for the shellfish food industry deserves further study. Emphasis is directed to the dinoflagellate species involved in their production, their chemical structures, and their specific mode of interaction with their principal natural molecular targets, the nicotinic acetylcholine receptors, or with the soluble acetylcholine-binding protein, used as a surrogate receptor model. The dinoflagellates Karenia selliformis and Alexandrium ostenfeldii / A. peruvianum have been implicated in the biosynthesis of gymnodimines and spirolides, while Vulcanodinium rugosum is the producer of pinnatoxins and portimine. The cyclic imine toxins are characterized by a macrocyclic skeleton comprising 14-27 carbon atoms, flanked by two conserved moieties, the cyclic imine and the spiroketal ring system. These phycotoxins generally display high affinity and broad specificity for the muscle type and neuronal nicotinic acetylcholine receptors, a feature consistent with their binding site at the receptor subunit interfaces, composed of residues highly conserved among all nAChRs, and explaining the diverse toxicity among animal species. This is an article for the special issue XVth International Symposium on Cholinergic Mechanisms.
Collapse
Affiliation(s)
- Jordi Molgó
- Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR 9197, Centre National de la Recherche Scientifique (CNRS)/Université Paris-Sud, Gif-sur-Yvette Cedex, France
| | - Pascale Marchot
- Aix-Marseille Université / Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques laboratory, Marseille, France
| | - Rómulo Aráoz
- Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR 9197, Centre National de la Recherche Scientifique (CNRS)/Université Paris-Sud, Gif-sur-Yvette Cedex, France
| | - Evelyne Benoit
- Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR 9197, Centre National de la Recherche Scientifique (CNRS)/Université Paris-Sud, Gif-sur-Yvette Cedex, France
| | - Bogdan I Iorga
- Centre National de la Recherche Scientifique (CNRS), Institut de Chimie des Substances Naturelles, UPR 2301, Labex LERMIT, Gif-sur-Yvette, France
| | - Armen Zakarian
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California, USA
| | - Palmer Taylor
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Yves Bourne
- Aix-Marseille Université / Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques laboratory, Marseille, France
| | - Denis Servent
- Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, Gif-sur-Yvette, France
| |
Collapse
|
20
|
Forebrain-selective AMPA-receptor antagonism guided by TARP γ-8 as an antiepileptic mechanism. Nat Med 2016; 22:1496-1501. [DOI: 10.1038/nm.4221] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
|
21
|
Tillman TS, Alvarez FJD, Reinert NJ, Liu C, Wang D, Xu Y, Xiao K, Zhang P, Tang P. Functional Human α7 Nicotinic Acetylcholine Receptor (nAChR) Generated from Escherichia coli. J Biol Chem 2016; 291:18276-82. [PMID: 27385587 DOI: 10.1074/jbc.m116.729970] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Indexed: 11/06/2022] Open
Abstract
Human Cys-loop receptors are important therapeutic targets. High-resolution structures are essential for rational drug design, but only a few are available due to difficulties in obtaining sufficient quantities of protein suitable for structural studies. Although expression of proteins in E. coli offers advantages of high yield, low cost, and fast turnover, this approach has not been thoroughly explored for full-length human Cys-loop receptors because of the conventional wisdom that E. coli lacks the specific chaperones and post-translational modifications potentially required for expression of human Cys-loop receptors. Here we report the successful production of full-length wild type human α7nAChR from E. coli Chemically induced chaperones promote high expression levels of well-folded proteins. The choice of detergents, lipids, and ligands during purification determines the final protein quality. The purified α7nAChR not only forms pentamers as imaged by negative-stain electron microscopy, but also retains pharmacological characteristics of native α7nAChR, including binding to bungarotoxin and positive allosteric modulators specific to α7nAChR. Moreover, the purified α7nAChR injected into Xenopus oocytes can be activated by acetylcholine, choline, and nicotine, inhibited by the channel blockers QX-222 and phencyclidine, and potentiated by the α7nAChR specific modulators PNU-120596 and TQS. The successful generation of functional human α7nAChR from E. coli opens a new avenue for producing mammalian Cys-loop receptors to facilitate structure-based rational drug design.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Xu
- From the Departments of Anesthesiology, Structural Biology, Pharmacology and Chemical Biology, and
| | | | | | - Pei Tang
- From the Departments of Anesthesiology, Pharmacology and Chemical Biology, and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
22
|
Couesnon A, Aráoz R, Iorga BI, Benoit E, Reynaud M, Servent D, Molgó J. The Dinoflagellate Toxin 20-Methyl Spirolide-G Potently Blocks Skeletal Muscle and Neuronal Nicotinic Acetylcholine Receptors. Toxins (Basel) 2016; 8:E249. [PMID: 27563924 PMCID: PMC5037475 DOI: 10.3390/toxins8090249] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/06/2016] [Accepted: 08/12/2016] [Indexed: 11/16/2022] Open
Abstract
The cyclic imine toxin 20-methyl spirolide G (20-meSPX-G), produced by the toxigenic dinoflagellate Alexandrium ostenfeldii/Alexandrium peruvianum, has been previously reported to contaminate shellfish in various European coastal locations, as revealed by mouse toxicity bioassay. The aim of the present study was to determine its toxicological profile and its molecular target selectivity. 20-meSPX-G blocked nerve-evoked isometric contractions in isolated mouse neuromuscular preparations, while it had no action on contractions elicited by direct electrical stimulation, and reduced reversibly nerve-evoked compound muscle action potential amplitudes in anesthetized mice. Voltage-clamp recordings in Xenopus oocytes revealed that 20-meSPX-G potently inhibited currents evoked by ACh on Torpedo muscle-type and human α7 nicotinic acetylcholine receptors (nAChR), whereas lower potency was observed in human α4β2 nAChR. Competition-binding assays showed that 20-meSPX-G fully displaced [³H]epibatidine binding to HEK-293 cells expressing the human α3β2 (Ki = 0.040 nM), whereas a 90-fold lower affinity was detected in human α4β2 nAChR. The spirolide displaced [(125)I]α-bungarotoxin binding to Torpedo membranes (Ki = 0.028 nM) and in HEK-293 cells expressing chick chimeric α7-5HT₃ nAChR (Ki = 0.11 nM). In conclusion, this is the first study to demonstrate that 20-meSPX-G is a potent antagonist of nAChRs, and its subtype selectivity is discussed on the basis of molecular docking models.
Collapse
MESH Headings
- Action Potentials
- Animals
- Binding Sites
- Binding, Competitive
- Bridged Bicyclo Compounds, Heterocyclic/metabolism
- Chickens
- Cholinergic Fibers/drug effects
- Cholinergic Fibers/metabolism
- Dose-Response Relationship, Drug
- Electric Stimulation
- Female
- HEK293 Cells
- Humans
- In Vitro Techniques
- Isometric Contraction/drug effects
- Mice
- Molecular Docking Simulation
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/innervation
- Muscle, Skeletal/metabolism
- Neuromuscular Junction/drug effects
- Neuromuscular Junction/metabolism
- Nicotinic Antagonists/chemistry
- Nicotinic Antagonists/metabolism
- Nicotinic Antagonists/toxicity
- Protein Binding
- Protein Conformation
- Pyridines/metabolism
- Receptors, Nicotinic/chemistry
- Receptors, Nicotinic/drug effects
- Receptors, Nicotinic/genetics
- Receptors, Nicotinic/metabolism
- Spiro Compounds/chemistry
- Spiro Compounds/metabolism
- Spiro Compounds/toxicity
- Structure-Activity Relationship
- Torpedo
- Transfection
- Xenopus laevis
Collapse
Affiliation(s)
- Aurélie Couesnon
- Institut des Neurosciences Paris-Saclay, UMR 9197 CNRS/Université Paris-Sud, F-91190 Gif-sur-Yvette, France.
| | - Rómulo Aráoz
- Institut des Neurosciences Paris-Saclay, UMR 9197 CNRS/Université Paris-Sud, F-91190 Gif-sur-Yvette, France.
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, bâtiment 152, F-91191 Gif-sur-Yvette, France.
| | - Bogdan I Iorga
- Centre National de la Recherche Scientifique (CNRS), Institut de Chimie des Substances Naturelles, UPR 2301, Labex LERMIT, F-91198 Gif-sur-Yvette, France.
| | - Evelyne Benoit
- Institut des Neurosciences Paris-Saclay, UMR 9197 CNRS/Université Paris-Sud, F-91190 Gif-sur-Yvette, France.
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, bâtiment 152, F-91191 Gif-sur-Yvette, France.
| | - Morgane Reynaud
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, bâtiment 152, F-91191 Gif-sur-Yvette, France.
| | - Denis Servent
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, bâtiment 152, F-91191 Gif-sur-Yvette, France.
| | - Jordi Molgó
- Institut des Neurosciences Paris-Saclay, UMR 9197 CNRS/Université Paris-Sud, F-91190 Gif-sur-Yvette, France.
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biologie et Technologies de Saclay (IBITECS), Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, bâtiment 152, F-91191 Gif-sur-Yvette, France.
| |
Collapse
|
23
|
Mazzo F, Zwart R, Serratto GM, Gardinier KM, Porter W, Reel J, Maraula G, Sher E. Reconstitution of synaptic Ion channels from rodent and human brain in Xenopus oocytes: a biochemical and electrophysiological characterization. J Neurochem 2016; 138:384-96. [PMID: 27216696 DOI: 10.1111/jnc.13675] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/10/2016] [Accepted: 05/18/2016] [Indexed: 11/29/2022]
Abstract
Disruption in the expression and function of synaptic proteins, and ion channels in particular, is critical in the pathophysiology of human neuropsychiatric and neurodegenerative diseases. However, very little is known regarding the functional and pharmacological properties of native synaptic human ion channels, and their potential changes in pathological conditions. Recently, an electrophysiological technique has been enabled for studying the functional and pharmacological properties of ion channels present in crude membrane preparation obtained from post-mortem frozen brains. We here extend these studies by showing that human synaptic ion channels also can be studied in this way. Synaptosomes purified from different regions of rodent and human brain (control and Alzheimer's) were characterized biochemically for enrichment of synaptic proteins, and expression of ion channel subunits. The same synaptosomes were also reconstituted in Xenopus oocytes, in which the functional and pharmacological properties of the native synaptic ion channels were characterized using the voltage clamp technique. We show that we can detect GABA, (RS)-α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and NMDA receptors, and modulate them pharmacologically with selective agonists, antagonists, and allosteric modulators. Furthermore, changes in ion channel expression and function were detected in synaptic membranes from Alzheimer's brains. Our present results demonstrate the possibility to investigate synaptic ion channels from healthy and pathological brains. This method of synaptosomes preparation and injection into oocytes is a significant improvement over the earlier method. It opens the way to directly testing, on native ion channels, the effects of novel drugs aimed at modulating important classes of synaptic targets. Disruption in the expression and function of synaptic ion channels is critical in the pathophysiology of human neurodegenerative diseases. We here show that synaptosomes purified from rodent and human frozen brain (control and Alzheimer disease) can be studied both biochemically and functionally. This method opens the way to directly testing the effects of novel drugs on native ion channels.
Collapse
Affiliation(s)
- Francesca Mazzo
- Lilly Research Centre, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, GU20 6HP, UK
| | - Ruud Zwart
- Lilly Research Centre, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, GU20 6HP, UK
| | - Giulia Maia Serratto
- Lilly Research Centre, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, GU20 6HP, UK
| | - Kevin M Gardinier
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Warren Porter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Jon Reel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Giovanna Maraula
- Lilly Research Centre, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, GU20 6HP, UK
| | - Emanuele Sher
- Lilly Research Centre, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, GU20 6HP, UK
| |
Collapse
|
24
|
Limon A, Estrada-Mondragón A, Ruiz JMR, Miledi R. Dipicrylamine Modulates GABAρ1 Receptors through Interactions with Residues in the TM4 and Cys-Loop Domains. Mol Pharmacol 2016; 89:446-56. [PMID: 26869399 DOI: 10.1124/mol.116.103432] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 02/09/2016] [Indexed: 02/02/2023] Open
Abstract
Dipicrylamine (DPA) is a commonly used acceptor agent in Förster resonance energy transfer experiments that allows the study of high-frequency neuronal activity in the optical monitoring of voltage in living cells. However, DPA potently antagonizes GABAA receptors that contain α1 and β2 subunits by a mechanism which is not clearly understood. In this work, we aimed to determine whether DPA modulation is a general phenomenon of Cys-loop ligand-gated ion channels (LGICs), and whether this modulation depends on particular amino acid residues. For this, we studied the effects of DPA on human homomeric GABAρ1, α7 nicotinic, and 5-HT3A serotonin receptors expressed in Xenopus oocytes. Our results indicate that DPA is an allosteric modulator of GABAρ1 receptors with an IC50 of 1.6 µM, an enhancer of α7 nicotinic receptors at relatively high concentrations of DPA, and has little, if any, effect on 5-HT3A receptors. DPA antagonism of GABAρ1 was strongly enhanced by preincubation, was slightly voltage-dependent, and its washout was accelerated by bovine serum albumin. These results indicate that DPA modulation is not a general phenomenon of LGICs, and structural differences between receptors may account for disparities in DPA effects. In silico modeling of DPA docking to GABAρ1, α7 nicotinic, and 5-HT3A receptors suggests that a hydrophobic pocket within the Cys-loop and the M4 segment in GABAρ1, located at the extracellular/membrane interface, facilitates the interaction with DPA that leads to inhibition of the receptor. Functional examinations of mutant receptors support the involvement of the M4 segment in the allosteric modulation of GABAρ1 by DPA.
Collapse
Affiliation(s)
- Agenor Limon
- Neurobiology and Behavior, University of California, Irvine, California (A.L., J.M.R.R., R.M.); Psychiatry and Human Behavior, University of California, Irvine, California (A.L.); and The Queensland Brain Institute, St. Lucia, Queensland, Australia (A.E.-M.)
| | - Argel Estrada-Mondragón
- Neurobiology and Behavior, University of California, Irvine, California (A.L., J.M.R.R., R.M.); Psychiatry and Human Behavior, University of California, Irvine, California (A.L.); and The Queensland Brain Institute, St. Lucia, Queensland, Australia (A.E.-M.)
| | - Jorge M Reyes Ruiz
- Neurobiology and Behavior, University of California, Irvine, California (A.L., J.M.R.R., R.M.); Psychiatry and Human Behavior, University of California, Irvine, California (A.L.); and The Queensland Brain Institute, St. Lucia, Queensland, Australia (A.E.-M.)
| | - Ricardo Miledi
- Neurobiology and Behavior, University of California, Irvine, California (A.L., J.M.R.R., R.M.); Psychiatry and Human Behavior, University of California, Irvine, California (A.L.); and The Queensland Brain Institute, St. Lucia, Queensland, Australia (A.E.-M.)
| |
Collapse
|
25
|
Acetylcholine receptors from human muscle as pharmacological targets for ALS therapy. Proc Natl Acad Sci U S A 2016; 113:3060-5. [PMID: 26929355 DOI: 10.1073/pnas.1600251113] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons that leads to progressive paralysis of skeletal muscle. Studies of ALS have revealed defects in expression of acetylcholine receptors (AChRs) in skeletal muscle that occur even in the absence of motor neuron anomalies. The endocannabinoid palmitoylethanolamide (PEA) modified the clinical conditions in one ALS patient, improving muscle force and respiratory efficacy. By microtransplanting muscle membranes from selected ALS patients into Xenopus oocytes, we show that PEA reduces the desensitization of acetylcholine-evoked currents after repetitive neurotransmitter application (i.e., rundown). The same effect was observed using muscle samples from denervated (non-ALS) control patients. The expression of human recombinant α1β1γδ (γ-AChRs) and α1β1εδ AChRs (ε-AChRs) in Xenopus oocytes revealed that PEA selectively affected the rundown of ACh currents in ε-AChRs. A clear up-regulation of the α1 subunit in muscle from ALS patients compared with that from non-ALS patients was found by quantitative PCR, but no differential expression was found for other subunits. Clinically, ALS patients treated with PEA showed a lower decrease in their forced vital capacity (FVC) over time as compared with untreated ALS patients, suggesting that PEA can enhance pulmonary function in ALS. In the present work, data were collected from a cohort of 76 ALS patients and 17 denervated patients. Our results strengthen the evidence for the role of skeletal muscle in ALS pathogenesis and pave the way for the development of new drugs to hamper the clinical effects of the disease.
Collapse
|
26
|
Bittolo T, Raminelli CA, Deiana C, Baj G, Vaghi V, Ferrazzo S, Bernareggi A, Tongiorgi E. Pharmacological treatment with mirtazapine rescues cortical atrophy and respiratory deficits in MeCP2 null mice. Sci Rep 2016; 6:19796. [PMID: 26806603 PMCID: PMC4726391 DOI: 10.1038/srep19796] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/14/2015] [Indexed: 01/22/2023] Open
Abstract
Loss of MeCP2 (Methyl CpG binding protein 2) in Rett syndrome (RTT) causes brain weight decrease, shrinkage of the cortex with reduced dendritic arborization, behavioral abnormalities, seizures and cardio-respiratory complications. The observed monoamine neurotransmitters reduction in RTT suggested antidepressants as a possible therapy. We treated MeCP2-null mice from postnatal-day 28 for two weeks with desipramine, already tested in RTT, or mirtazapine, an antidepressant with limited side-effects, known to promote GABA release. Mirtazapine was more effective than desipramine in restoring somatosensory cortex thickness by fully rescuing pyramidal neurons dendritic arborization and spine density. Functionally, mirtazapine treatment normalized heart rate, breath rate, anxiety levels, and eliminated the hopping behavior observed in MeCP2-null mice, leading to improved phenotypic score. These morphological and functional effects of mirtazapine were accompanied by reestablishment of the GABAergic and glutamatergic receptor activity recorded in cortex and brainstem tissues. Thus, mirtazapine can represent a new potential pharmacological treatment for the Rett syndrome.
Collapse
Affiliation(s)
- Tamara Bittolo
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Carlo Antonio Raminelli
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Chiara Deiana
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Gabriele Baj
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Valentina Vaghi
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Sara Ferrazzo
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Via L. Giorgieri, 5-34127 Trieste, Italy
| |
Collapse
|
27
|
Crespin L, Legros C, List O, Tricoire-Leignel H, Mattei C. Injection of insect membrane in Xenopus oocyte: An original method for the pharmacological characterization of neonicotinoid insecticides. J Pharmacol Toxicol Methods 2015; 77:10-6. [PMID: 26391340 DOI: 10.1016/j.vascn.2015.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Insect nicotinic acetylcholine receptors (nAChRs) represent a major target of insecticides, belonging to the neonicotinoid family. However, the pharmacological profile of native nAChRs is poorly documented, mainly because of a lack of knowledge of their subunit stoichiometry, their tissue distribution and the weak access to nAChR-expressing cells. In addition, the expression of insect nAChRs in heterologous systems remains hard to achieve. Therefore, the structure-activity characterization of nAChR-targeting insecticides is made difficult. The objective of the present study was to characterize insect nAChRs by an electrophysiological approach in a heterologous system naturally devoid of these receptors to allow a molecular/cellular investigation of the mode of action of neonicotinoids. Methods To overcome impediments linked to the expression of insect nAChR mRNA or cDNA, we chose to inject insect membranes from the pea aphid (Acyrthosiphon pisum) into Xenopus oocytes. This microtransplantation technique was designed to gain access to native nAChRs embedded in their membrane, through direct stimulation with nicotinic agonists. Results We provide evidence that an enriched-nAChR membrane allows us to characterize native receptors. The presence of such receptors was confirmed with fluorescent α-BgTX labeling. Electrophysiological recordings of nicotine-induced inward currents allowed us to challenge the presence of functional nAChR. We compared the effect of nicotine (NIC) with clothianidin (CLO) and we assessed the effect of thiamethoxam (TMX). Discussion This technique has been recently highlighted with mammalian and human material as a powerful functional approach, but has, to our knowledge, never been used with insect membrane. In addition, the use of the insect membrane microtransplantation opens a new and original way for pharmacological screening of neurotoxic insecticides, including neonicotinoids. Moreover, it might also be a powerful tool to investigate the pharmacological properties of insect nAChR.
Collapse
Affiliation(s)
- Lucille Crespin
- Laboratoire de Biologie Neurovasculaire et Mitochondriale Intégrée CNRS UMR6214, INSERM U1083, Univ. Angers Faculté de Médecine, rue Haute de Reculée, 49045 Angers cedex 01, France
| | - Christian Legros
- Laboratoire de Biologie Neurovasculaire et Mitochondriale Intégrée CNRS UMR6214, INSERM U1083, Univ. Angers Faculté de Médecine, rue Haute de Reculée, 49045 Angers cedex 01, France
| | - Olivier List
- Récepteurs et Canaux Ioniques Membranaires, UPRES-EA 2647 USC INRA 1330, SFR 4207 QUASAV, Univ. Angers, 2 Bd Lavoisier, 49045 Angers cedex 01, France
| | - Hélène Tricoire-Leignel
- INRA/Université d'Angers, Neuroéthologie-RCIM, UPRES-EA 2647 USC INRA 1330, SFR 4207 QUASAV, 42, rue Georges Morel, 49071 Beaucouzé, France.
| | - César Mattei
- Laboratoire de Biologie Neurovasculaire et Mitochondriale Intégrée CNRS UMR6214, INSERM U1083, Univ. Angers Faculté de Médecine, rue Haute de Reculée, 49045 Angers cedex 01, France.
| |
Collapse
|
28
|
Candelario M, Cuellar E, Reyes-Ruiz JM, Darabedian N, Feimeng Z, Miledi R, Russo-Neustadt A, Limon A. Direct evidence for GABAergic activity of Withania somnifera on mammalian ionotropic GABAA and GABAρ receptors. JOURNAL OF ETHNOPHARMACOLOGY 2015; 171:264-72. [PMID: 26068424 DOI: 10.1016/j.jep.2015.05.058] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/22/2015] [Accepted: 05/30/2015] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Withania somnifera (WS) has been traditionally used in Ayurvedic medicine as a remedy for debility, stress, nervous exhaustion, insomnia, loss of memory, and to enhance cognitive function. This study provides an empirical evidence to support the traditional use of WS to aid in mental process engaging GABAergic signaling. AIM OF THE STUDY We evaluated the effect of aqueous WS root extract (aqWS), and its two main components, withaferin A and withanolide A, on the main inhibitory receptors in the central nervous system: ionotropic GABAA receptors. MATERIALS AND METHODS The pharmacological activity of aqWS, withaferin A and withanolide A, was tested on native rat brain GABAA channels microtransplanted into Xenopus oocytes and GABAρ1 receptors heterologously expressed in oocytes. The GABAergic activity of aqWS compounds was evaluated by the two-electrode voltage-clamp method and the fingerprint of the extract was done by LC-MS. RESULTS Concentration-dependent inward ion currents were elicited by aqWS in microtransplanted oocytes with an EC50 equivalent to 4.7 mg/mL and a Hill coefficient (nH) of 1.6. The GABAA receptor antagonist bicuculline blocked these currents. Our results show that aqWS activated inotropic GABAA channels but with lower efficacy compared to the endogenous agonist GABA. We also demonstrate for first time that aqWS is a potent agonist of GABAρ1 receptors. GABAρ1 receptors were 27 fold more sensitive to aqWS than GABAA receptors. Furthermore, aqWS activated GABAρ1 receptors eliciting maximum currents that were no significantly different to those produced by GABA (paired t-test; p=0.533). The differential activity on GABAA and GABA ρ1 receptors and the reported lack of significant GABA presence in WS root extract indicates that the GABAergic activity of aqWS is not mediated by GABA. WS main active components, witaferin A and withanolide A, were tested to determine if they were responsible for the activation of the GABA receptors. Neither compound activated GABAA nor GABAρ1 receptors, suggesting that other constituent/s in WS are responsible for GABAA receptor mediated responses. CONCLUSIONS Our results provide evidence indicating that key constituents in WS may have an important role in the development of pharmacological treatments for neurological disorders associated with GABAergic signaling dysfunction such as general anxiety disorders, sleep disturbances, muscle spasms, and seizures. In addition, the differential activation of GABA receptor subtypes elucidates a potential mechanism by which WS accomplishes its reported adaptogenic properties.
Collapse
Affiliation(s)
- Manuel Candelario
- Biological Sciences Department, California State University, Los Angeles, CA, United States
| | - Erika Cuellar
- Biological Sciences Department, California State University, Los Angeles, CA, United States
| | | | - Narek Darabedian
- Department of Chemistry and Biochemistry, California State University, Los Angeles, CA, United States
| | - Zhou Feimeng
- Department of Chemistry and Biochemistry, California State University, Los Angeles, CA, United States
| | - Ricardo Miledi
- Department of Neurobiology & Behavior, University of California, Irvine, CA, United States
| | - Amelia Russo-Neustadt
- Biological Sciences Department, California State University, Los Angeles, CA, United States
| | - Agenor Limon
- Department of Neurobiology & Behavior, University of California, Irvine, CA, United States; Department of Psychiatry and Human Behavior, University of California, Irvine, CA, United States.
| |
Collapse
|
29
|
Roseti C, van Vliet EA, Cifelli P, Ruffolo G, Baayen JC, Di Castro MA, Bertollini C, Limatola C, Aronica E, Vezzani A, Palma E. GABAA currents are decreased by IL-1β in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis. Neurobiol Dis 2015; 82:311-320. [PMID: 26168875 DOI: 10.1016/j.nbd.2015.07.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most prevalent form of adult focal onset epilepsy often associated with drug-resistant seizures. Numerous studies suggest that neuroinflammatory processes are pathologic hallmarks of both experimental and human epilepsy. In particular, the interleukin (IL)-1β/IL-1 receptor type 1 (R1) axis is activated in epileptogenic tissue, where it contributes significantly to the generation and recurrence of seizures in animal models. In this study, we investigated whether IL-1β affects the GABA-evoked currents (I(GABA)) in TLE tissue from humans. Given the limited availability of fresh human brain specimens, we used the "microtransplantation" method of injecting Xenopus oocytes with membranes from surgically resected hippocampal and cortical tissue from 21 patients with TLE and hippocampal sclerosis (HS), hippocampal tissue from five patients with TLE without HS, and autoptic and surgical brain specimens from 15 controls without epilepsy. We report the novel finding that pathophysiological concentrations of IL-1β decreased the I(GABA) amplitude by up to 30% in specimens from patients with TLE with or without HS, but not in control tissues. This effect was reproduced by patch-clamp recordings on neurons in entorhinal cortex slices from rats with chronic epilepsy, and was not observed in control slices. In TLE specimens from humans, the IL-1β effect was mediated by IL-1R1 and PKC. We also showed that IL-1R1 and IRAK1, the proximal kinase mediating the IL-1R1 signaling, are both up-regulated in the TLE compared with control specimens, thus supporting the idea that the IL-1β/IL-R1 axis is activated in human epilepsy. Our findings suggest a novel mechanism possibly underlying the ictogenic action of IL-1β, thus suggesting that this cytokine contributes to seizure generation in human TLE by reducing GABA-mediated neurotransmission.
Collapse
Affiliation(s)
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Pierangelo Cifelli
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy; Ri.MED Foundation, Palermo, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Johannes C Baayen
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Maria Amalia Di Castro
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Cristina Bertollini
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN-Heemstede), The Netherlands
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy.
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy.
| |
Collapse
|
30
|
Aráoz R, Ouanounou G, Iorga BI, Goudet A, Alili D, Amar M, Benoit E, Molgó J, Servent D. The Neurotoxic Effect of 13,19-Didesmethyl and 13-Desmethyl Spirolide C Phycotoxins Is Mainly Mediated by Nicotinic Rather Than Muscarinic Acetylcholine Receptors. Toxicol Sci 2015; 147:156-67. [DOI: 10.1093/toxsci/kfv119] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Bernareggi A, Ren E, Borelli V, Vita F, Constanti A, Zabucchi G. Xenopus laevis Oocytes as a Model System for Studying the Interaction Between Asbestos Fibres and Cell Membranes. Toxicol Sci 2015; 145:263-72. [PMID: 25745069 DOI: 10.1093/toxsci/kfv050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The mode of interaction of asbestos fibres with cell membranes is still debatable. One reason is the lack of a suitable and convenient cellular model to investigate the causes of asbestos toxicity. We studied the interaction of asbestos fibres with Xenopus laevis oocytes, using electrophysiological and morphological methods. Oocytes are large single cells, with a limited ability to endocytose molecular ligands; we therefore considered these cells to be a good model for investigating the nature of asbestos/membrane interactions. Electrophysiological recordings were performed to compare the passive electrical membrane properties, and those induced by applying positive or negative voltage steps, in untreated oocytes and those exposed to asbestos fibre suspensions. Ultrastructural analysis visualized in detail, any morphological changes of the surface membrane caused by the fibre treatment. Our results demonstrate that Amosite and Crocidolite-type asbestos fibres significantly modify the properties of the membrane, starting soon after exposure. Cells were routinely depolarized, their input resistance decreased, and the slow outward currents evoked by step depolarizations were dramatically enhanced. Reducing the availability of surface iron contained in the structure of the fibres with cation chelators, abolished these effects. Ultrastructural analysis of the fibre-exposed oocytes showed no evidence of phagocytic events. Our results demonstrate that asbestos fibres modify the oocyte membrane, and we propose that these cells represent a viable model for studying the asbestos/cell membrane interaction. Our findings also open the possibly for finding specific competitors capable of hindering the asbestos-cell membrane interaction as a means of tackling the long-standing asbestos toxicity problem.
Collapse
Affiliation(s)
- Annalisa Bernareggi
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| | - Elisa Ren
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| | - Violetta Borelli
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| | - Francesca Vita
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| | - Andrew Constanti
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| | - Giuliano Zabucchi
- *Department of Life Sciences; Centre for Neuroscience B.R.A.I.N., University of Trieste, 34127 Trieste, Italy; and Department of Pharmacology, UCL School of Pharmacy, London, UK WC1N 1AX
| |
Collapse
|
32
|
Zwart R, Sher E, Ping X, Jin X, Sims JR, Chappell AS, Gleason SD, Hahn PJ, Gardinier K, Gernert DL, Hobbs J, Smith JL, Valli SN, Witkin JM. Perampanel, an antagonist of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, for the treatment of epilepsy: studies in human epileptic brain and nonepileptic brain and in rodent models. J Pharmacol Exp Ther 2014; 351:124-33. [PMID: 25027316 DOI: 10.1124/jpet.114.212779] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Perampanel [Fycompa, 2-(2-oxo-1-phenyl-5-pyridin-2-yl-1,2-dihydropyridin-3-yl)benzonitrile hydrate 4:3; Eisai Inc., Woodcliff Lake, NJ] is an AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor antagonist used as an adjunctive treatment of partial-onset seizures. We asked whether perampanel has AMPA receptor antagonist activity in both the cerebral cortex and hippocampus associated with antiepileptic efficacy and also in the cerebellum associated with motor side effects in rodent and human brains. We also asked whether epileptic or nonepileptic human cortex is similarly responsive to AMPA receptor antagonism by perampanel. In rodent models, perampanel decreased epileptic-like activity in multiple seizure models. However, doses of perampanel that had anticonvulsant effects were within the same range as those engendering motor side effects. Perampanel inhibited native rat and human AMPA receptors from the hippocampus as well as the cerebellum that were reconstituted into Xenopus oocytes. In addition, with the same technique, we found that perampanel inhibited AMPA receptors from hippocampal tissue that had been removed from a patient who underwent surgical resection for refractory epilepsy. Perampanel inhibited AMPA receptor-mediated ion currents from all the tissues investigated with similar potency (IC50 values ranging from 2.6 to 7.0 μM). Cortical slices from the left temporal lobe derived from the same patient were studied in a 60-microelectrode array. Large field potentials were evoked on at least 45 channels of the array, and 10 μM perampanel decreased their amplitude and firing rate. Perampanel also produced a 33% reduction in the branching parameter, demonstrating the effects of perampanel at the network level. These data suggest that perampanel blocks AMPA receptors globally across the brain to account for both its antiepileptic and side-effect profile in rodents and epileptic patients.
Collapse
Affiliation(s)
- R Zwart
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - E Sher
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - X Ping
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - X Jin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - J R Sims
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - A S Chappell
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - S D Gleason
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - P J Hahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - K Gardinier
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - D L Gernert
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - J Hobbs
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - J L Smith
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - S N Valli
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| | - J M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (J.R.S., A.S.C., S.D.G., P.J.H., K.G., D.L.G., S.N.V., J.M.W.); Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, United Kingdom (R.Z., E.S.); and Indiana University/Purdue University, Riley Hospital, Indianapolis, Indiana (X.P., X.J., J.H., J.L.S.)
| |
Collapse
|
33
|
Roseti C, Fucile S, Lauro C, Martinello K, Bertollini C, Esposito V, Mascia A, Catalano M, Aronica E, Limatola C, Palma E. Fractalkine/CX3CL1 modulates GABAA currents in human temporal lobe epilepsy. Epilepsia 2013; 54:1834-44. [PMID: 24032743 DOI: 10.1111/epi.12354] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2013] [Indexed: 11/30/2022]
Abstract
PURPOSE The chemokine fractalkine/CX3CL1 and its receptor CX3CR1 are widely expressed in the central nervous system (CNS). Recent evidence showed that CX3CL1 participates in inflammatory responses that are common features of CNS disorders, such as epilepsy. Mesial temporal lobe epilepsy (MTLE) is the prevalent form of focal epilepsy in adults, and hippocampal sclerosis (HS) represents the most common underlying pathologic abnormality, as demonstrated at autopsy and postresection studies. Relevant features of MTLE are a characteristic pattern of neuronal loss, as are astrogliosis and microglia activation. Several factors affect epileptogenesis in patients with MTLE, including a lack of γ-aminobutyric acid (GABA)ergic inhibitory efficacy. Therefore, experiments were designed to investigate whether, in MTLE brain tissues, CX3CL1 may influence GABAA receptor (GABAA R) mediated transmission, with a particular focus on the action of CX3CL1 on the use-dependent decrease (rundown) of the GABA-evoked currents (IGABA ), a feature underlying the reduction of GABAergic function in epileptic tissue. METHODS Patch-clamp recordings were obtained from cortical pyramidal neurons in slices from six MTLE patients after surgery. Alternatively, the cell membranes from epileptic brain tissues of 17 MTLE patients or from surgical samples and autopsies of nonepileptic patients were microtransplanted into Xenopus oocytes, and IGABA were recorded using the standard two-microelectrode voltage-clamp technique. Immunohistochemical staining and double-labeling studies were carried out on the same brain tissues to analyze CX3CR1 expression. KEY FINDINGS In native pyramidal neurons from cortical slices of patients with MTLE, CX3CL1 reduced IGABA rundown and affected the recovery of IGABA amplitude from rundown. These same effects were confirmed in oocytes injected with cortical and hippocampal MTLE membranes, whereas CX3CL1 did not influence IGABA in oocytes injected with nonepileptic tissues. Consistent with a specific effect of CX3CL1 on tissues from patients with MTLE, CX3CR1 immunoreactivity was higher in MTLE sclerotic hippocampi than in control tissues, with a prominent expression in activated microglial cells. SIGNIFICANCE These findings indicate a role for CX3CL1 in MTLE, supporting recent evidence on the relevance of brain inflammation in human epilepsies. Our data demonstrate that in MTLE tissues the reduced GABAergic function can be modulated by CX3CL1. The increased CX3CR1 expression in microglia and the modulation by CX3CL1 of GABAergic currents in human epileptic brain suggests new therapeutic approaches for drug-resistant epilepsies based on the evidence that the propagation of seizures can be influenced by inflammatory processes.
Collapse
|
34
|
Molgó J, Aráoz R, Benoit E, Iorga BI. Physical and virtual screening methods for marine toxins and drug discovery targeting nicotinic acetylcholine receptors. Expert Opin Drug Discov 2013; 8:1203-23. [DOI: 10.1517/17460441.2013.822365] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Cifelli P, Palma E, Roseti C, Verlengia G, Simonato M. Changes in the sensitivity of GABAA current rundown to drug treatments in a model of temporal lobe epilepsy. Front Cell Neurosci 2013; 7:108. [PMID: 23874269 PMCID: PMC3708152 DOI: 10.3389/fncel.2013.00108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/21/2013] [Indexed: 01/26/2023] Open
Abstract
The pharmacological treatment of mesial temporal lobe epilepsy (mTLE), the most common epileptic syndrome in adults, is still unsatisfactory, as one-third of the patients are or become refractory to antiepileptic agents. Refractoriness may depend upon drug-induced alterations, but the disease per se may also undergo a progressive evolution that affects the sensitivity to drugs. mTLE has been shown to be associated with a dysfunction of the inhibitory signaling mediated by GABAA receptors. In particular, the repetitive activation of GABAA receptors produces a use-dependent decrease (rundown) of the evoked currents (IGABA), which is markedly enhanced in the hippocampus and cortex of drug-resistant mTLE patients. This phenomenon has been also observed in the pilocarpine model, where the increased IGABA rundown is observed in the hippocampus at the time of the first spontaneous seizure, then extends to the cortex and remains constant in the chronic phase of the disease. Here, we examined the sensitivity of IGABA to pharmacological modulation. We focused on the antiepileptic agent levetiracetam (LEV) and on the neurotrophin brain-derived neurotrophic factor (BDNF), which were previously reported to attenuate mTLE-induced increased rundown in the chronic human tissue. In the pilocarpine model, BDNF displayed a paramount effect, decreasing rundown in the hippocampus at the time of the first seizure, as well as in the hippocampus and cortex in the chronic period. In contrast, LEV did not affect rundown in the hippocampus, but attenuated it in the cortex. Interestingly, this effect of LEV was also observed on the still unaltered rundown observed in the cortex at the time of the first spontaneous seizure. These data suggest that the sensitivity of GABAA receptors to pharmacological interventions undergoes changes during the natural history of mTLE, implicating that the site of seizure initiation and the timing of treatment may highly affect the therapeutic outcome.
Collapse
Affiliation(s)
- Pierangelo Cifelli
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara Ferrara, Italy ; National Institute of Neuroscience Ferrara, Italy ; Ri.MED Foundation Palermo, Italy
| | | | | | | | | |
Collapse
|
36
|
Ochoa-de la Paz LD, Espino-Saldaña AE, Arellano-Ostoa R, Reyes JP, Miledi R, Martinez-Torres A. Characterization of an outward rectifying chloride current of Xenopus tropicalis oocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1743-53. [PMID: 23524227 DOI: 10.1016/j.bbamem.2013.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 03/08/2013] [Accepted: 03/13/2013] [Indexed: 12/19/2022]
Abstract
Here, we describe an outward rectifying current in Xenopus tropicalis oocytes that we have called xtClC-or. The current has two components; the major component is voltage activated and independent of intracellular or extracellular Ca(2+), whereas the second is a smaller component that is Ca(2+) dependent. The properties of the Ca(2+)-independent current, such as voltage dependence and outward rectification, resemble those of ClC anion channels/transporters. This current is sensitive to NPPB and NFA, insensitive to 9AC and DIDS, and showed a whole-cell conductance sequence of SCN(-)>I(-)>Br(-)>CI(-). RT-PCR revealed the expression in oocytes of ClC-2 to ClC-7, and major reductions of current amplitudes were observed when a ClC-5 antisense oligonucleotide was injected into oocytes. The Ca(2+)-dependent component was abated after injection of 10mM BAPTA or EGTA, whereas 10mMMg(2+) inhibited the current to 26±3.1%. This component was blocked by 9-AC, NFA, and NPPB, whereas DIDS did not elicit any evident effect. The ion sequence selectivity was SCN=I(-)>Br(-)>Cl(-). To try to determine the molecular identity that gives rise to this component we assessed by RT-PCR the expression of the Ca(2+)-dependent Cl(-) channel TMEM16A, which was found to be present in the oocytes. However, injection of antisense TMEM16A oligonucleotides did not inhibit the transient outward current. This result fits well with the electrophysiological data. Together, these results suggest that ClC-5 is a major, but not the sole channel responsible for this outwardly rectifying Cl(-) current.
Collapse
Affiliation(s)
- Lenin David Ochoa-de la Paz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus UNAM Juriquilla, Mexico
| | | | | | | | | | | |
Collapse
|
37
|
Conti L, Limon A, Palma E, Miledi R. Microtransplantation of cellular membranes from squid stellate ganglion reveals ionotropic GABA receptors. THE BIOLOGICAL BULLETIN 2013; 224:47-52. [PMID: 23493508 DOI: 10.1086/bblv224n1p47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The squid has been the most studied cephalopod, and it has served as a very useful model for investigating the events associated with nerve impulse generation and synaptic transmission. While the physiology of squid giant axons has been extensively studied, very little is known about the distribution and function of the neurotransmitters and receptors that mediate inhibitory transmission at the synapses. In this study we investigated whether γ-aminobutyric acid (GABA) activates neurotransmitter receptors in stellate ganglia membranes. To overcome the low abundance of GABA-like mRNAs in invertebrates and the low expression of GABA in cephalopods, we used a two-electrode voltage clamp technique to determine if Xenopus laevis oocytes injected with cell membranes from squid stellate ganglia responded to GABA. Using this method, membrane patches containing proteins and ion channels from the squid's stellate ganglion were incorporated into the surface of oocytes. We demonstrated that GABA activates membrane receptors in cellular membranes isolated from squid stellate ganglia. Using the same approach, we were able to record native glutamate-evoked currents. The squid's GABA receptors showed an EC(50) of 98 μmol l(-1) to GABA and were inhibited by zinc (IC(50) = 356 μmol l(-1)). Interestingly, GABA receptors from the squid were only partially blocked by bicuculline. These results indicate that the microtransplantation of native cell membranes is useful to identify and characterize scarce membrane proteins. Moreover, our data also support the role of GABA as an ionotropic neurotransmitter in cephalopods, acting through chloride-permeable membrane receptors.
Collapse
Affiliation(s)
- Luca Conti
- Grass Laboratory at the Marine Biological Laboratory, 7 MBL St., Woods Hole, MA 02543, USA.
| | | | | | | |
Collapse
|
38
|
Deflorio C, Palma E, Conti L, Roseti C, Manteca A, Giacomelli E, Catalano M, Limatola C, Inghilleri M, Grassi F. Riluzole blocks human muscle acetylcholine receptors. J Physiol 2012; 590:2519-28. [PMID: 22431338 DOI: 10.1113/jphysiol.2012.230201] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Riluzole, the only drug available against amyotrophic lateral sclerosis (ALS), has recently been shown to block muscle ACh receptors (AChRs), raising concerns about possible negative side-effects on neuromuscular transmission in treated patients. In this work we studied riluzole's impact on the function of muscle AChRs in vitro and on neuromuscular transmission in ALS patients, using electrophysiological techniques. Human recombinant AChRs composed of α(1)β(1)δ subunits plus the γ or ε subunit (γ- or ε-AChR) were expressed in HEK cells or Xenopus oocytes. In both preparations, riluzole at 0.5 μm, a clinically relevant concentration, reversibly reduced the amplitude and accelerated the decay of ACh-evoked current if applied before coapplication with ACh. The action on γ-AChRs was more potent and faster than on ε-AChRs. In HEK outside-out patches, riluzole-induced block of macroscopic ACh-evoked current gradually developed during the initial milliseconds of ACh presence. Single channel recordings in HEK cells and in human myotubes from ALS patients showed that riluzole prolongs channel closed time, but has no effect on channel conductance and open duration. Finally, compound muscle action potentials (CMAPs) evoked by nerve stimulation in ALS patients remained unaltered after a 1 week suspension of riluzole treatment. These data indicate that riluzole, while apparently safe with regard to synaptic transmission, may affect the function of AChRs expressed in denervated muscle fibres of ALS patients, with biological consequences that remain to be investigated.
Collapse
Affiliation(s)
- Cristina Deflorio
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Physiological characterization of human muscle acetylcholine receptors from ALS patients. Proc Natl Acad Sci U S A 2011; 108:20184-8. [PMID: 22128328 DOI: 10.1073/pnas.1117975108] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by progressive degeneration of motor neurons leading to muscle paralysis. Research in transgenic mice suggests that the muscle actively contributes to the disease onset, but such studies are difficult to pursue in humans and in vitro models would represent a good starting point. In this work we show that tiny amounts of muscle from ALS or from control denervated muscle, obtained by needle biopsy, are amenable to functional characterization by two different technical approaches: "microtransplantation" of muscle membranes into Xenopus oocytes and culture of myogenic satellite cells. Acetylcholine (ACh)-evoked currents and unitary events were characterized in oocytes and multinucleated myotubes. We found that ALS acetylcholine receptors (AChRs) retain their native physiological characteristics, being activated by ACh and nicotine and blocked by α-bungarotoxin (α-BuTX), d-tubocurarine (dTC), and galantamine. The reversal potential of ACh-evoked currents and the unitary channel behavior were also typical of normal muscle AChRs. Interestingly, in oocytes injected with muscle membranes derived from ALS patients, the AChRs showed a significant decrease in ACh affinity, compared with denervated controls. Finally, riluzole, the only drug currently used against ALS, reduced, in a dose-dependent manner, the ACh-evoked currents, indicating that its action remains to be fully characterized. The two methods described here will be important tools for elucidating the role of muscle in ALS pathogenesis and for developing drugs to counter the effects of this disease.
Collapse
|
40
|
Conti L, Palma E, Roseti C, Lauro C, Cipriani R, de Groot M, Aronica E, Limatola C. Anomalous levels of Cl- transporters cause a decrease of GABAergic inhibition in human peritumoral epileptic cortex. Epilepsia 2011; 52:1635-44. [PMID: 21635237 DOI: 10.1111/j.1528-1167.2011.03111.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Several factors contribute to epileptogenesis in patients with brain tumors, including reduced γ-aminobutyric acid (GABA)ergic inhibition. In particular, changes in Cl(-) homeostasis in peritumoral microenvironment, together with alterations of metabolism, are key processes leading to epileptogenesis in patients afflicted by glioma. It has been recently proposed that alterations of Cl(-) homeostasis could be involved in tumor cell migration and metastasis formation. In neurons, the regulation of intracellular Cl(-) concentration ([Cl(-) ](i) ) is mediated by NKCC1 and KCC2 transporters: NKCC1 increases while KCC2 decreases [Cl(-) ](i) . Experiments were thus designed to investigate whether, in human epileptic peritumoral cortex, alterations in the balance of NKCC1 and KCC2 activity may decrease the hyperpolarizing effects of GABA, thereby contributing to epileptogenesis in human brain tumors. METHODS Membranes from peritumoral cortical tissues of epileptic patients afflicted by gliomas (from II to IV WHO grade) and from cortical tissues of nonepileptic patients were injected into Xenopus oocytes leading to the incorporation of functional GABA(A) receptors. The GABA-evoked currents were recorded using standard two-microelectrode voltage-clamp technique. In addition, immunoblot analysis and immunohistochemical staining were carried out on membranes and tissues from the same patients. KEY FINDINGS We found that in oocytes injected with epileptic peritumoral cerebral cortex, the GABA-evoked currents had a more depolarized reversal potential (E(GABA) ) compared to those from nonepileptic healthy cortex. This difference of E(GABA) was abolished by the NKCC1 blocker bumetanide or unblocking of KCC2 with the Zn(2+) chelator TPEN. Moreover, Western blot analysis revealed an increased expression of NKCC1, and more modestly, of KCC2 transporters in epileptic peritumoral tissues compared to nonepileptic control tissues. In addition, NKCC1 immunoreactivity was strongly increased in peritumoral cortex with respect to nonepileptic cortex, with a prominent expression in neuronal cells. SIGNIFICANCE We report that the positive shift of E(GABA) in epileptic peritumoral human cortex is due to an altered expression of NKCC1 and KCC2, perturbing Cl(-) homeostasis, which might lead to a consequent reduction in GABAergic inhibition. These findings point to a key role of Cl(-) transporters KCC2 and NKCC1 in tumor-related epilepsy, suggesting a more specific drug therapy and surgical approaches for the epileptic patients afflicted by brain tumors.
Collapse
Affiliation(s)
- Luca Conti
- Pasteur Institute-Cenci Bolognetti Foundation, Department of Physiology and Pharmacology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Li G, Yang K, Zheng C, Liu Q, Chang Y, Kerrigan JF, Wu J. Functional rundown of gamma-aminobutyric acid(A) receptors in human hypothalamic hamartomas. Ann Neurol 2011; 69:664-72. [PMID: 21391233 DOI: 10.1002/ana.22298] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 08/23/2010] [Accepted: 09/17/2010] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Human hypothalamic hamartomas (HHs) are highly associated with treatment-resistant gelastic seizures. HHs are intrinsically epileptogenic, although the basic cellular mechanisms responsible for seizure activity are unknown. Altered gamma-aminobutyric acid (GABA) function can contribute to epileptogenesis in humans and animal models. Recently, functional GABA(A) receptor (GABA(A) R) rundown has been described in surgically resected human temporal lobe epilepsy tissue. We asked whether functional GABA(A) R rundown also occurs in human HH neurons. METHODS GABA(A) R-mediated currents were measured using perforated patch-clamp recordings in single neurons acutely dissociated from surgically resected HH tissue. In addition, functional GABA(A) Rs were expressed in Xenopus oocytes after microinjection with membrane fractions from either HH or control hypothalamus, and were studied with 2-electrode voltage-clamp recordings. RESULTS Perforated patch-clamp recordings in dissociated HH neurons showed that repetitive exposure to GABA (5 consecutive exposures to 0.1 mM GABA with 1-second duration and at 20-second intervals) induced a time-dependent rundown of whole-cell currents in small HH neurons, whereas large HH neurons showed much less rundown using the same protocol. Functional rundown was not observed in HH neurons with repetitive exposure to glycine or glutamate. Two-electrode voltage-clamp recordings (6 consecutive exposures to 1 mM GABA with 10-second duration and at 40-second intervals) induced GABA current rundown in Xenopus oocytes microinjected with HH membrane proteins, but not in the oocytes expressing hypothalamic membrane proteins derived from human autopsy controls. Functional rundown of GABA currents was significantly attenuated by intracellular application of adenosine triphosphate or the nonspecific phosphatase inhibitor, okadaic acid. INTERPRETATION Neurons from surgically resected human HH demonstrate functional rundown of GABA(A) R-mediated transmembrane currents in response to GABA agonist exposure. Rundown may be a marker for impaired GABAergic function and a contributing mechanism for seizure genesis within HH tissue.
Collapse
Affiliation(s)
- Guohui Li
- Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Bernareggi A, Reyes-Ruiz JM, Lorenzon P, Ruzzier F, Miledi R. Microtransplantation of acetylcholine receptors from normal or denervated rat skeletal muscles to frog oocytes. J Physiol 2011; 589:1133-42. [PMID: 21224230 DOI: 10.1113/jphysiol.2010.202994] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Cell membranes, carrying neurotransmitter receptors and ion channels, can be 'microtransplanted' into frog oocytes. This technique allows a direct functional characterization of the original membrane proteins, together with any associated molecules they may have, still embedded in their natural lipid environment. This approach has been previously demonstrated to be very useful to study neurotransmitter receptors and ion channels contained in cell membranes isolated from human brains. Here, we examined the possibility of using the microtransplantation method to study acetylcholine receptors from normal and denervated rat skeletal muscles. We found that the muscle membranes, carrying their fetal or adult acetylcholine receptor isoforms, could be efficiently microtransplanted to the oocyte membrane, making the oocytes become sensitive to acetylcholine. These results show that oocytes injected with skeletal muscle membranes efficiently incorporate functional acetylcholine receptors, thus making the microtransplantation approach a valuable tool to further investigate receptors and ion channels of human muscle diseases.
Collapse
Affiliation(s)
- Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, Via A. Fleming 22, I-34127 Trieste, Italy.
| | | | | | | | | |
Collapse
|
43
|
Abstract
Xenopus oocytes are a versatile expression system particularly suited for membrane transporters and channels. Oocytes have little background activity and therefore offer a very high signal-to-noise ratio for transporter and channel characterization. This chapter provides an overview of the basic methods used for the analysis of membrane transporters in this system, including preparation of oocytes, assays of transport activity, protocols for immunostaining and fluorescence microscopy, and other assays to study surface expression.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Canberra, Australia.
| |
Collapse
|
44
|
Enhancement of GABA(A)-current run-down in the hippocampus occurs at the first spontaneous seizure in a model of temporal lobe epilepsy. Proc Natl Acad Sci U S A 2010; 107:3180-5. [PMID: 20133704 DOI: 10.1073/pnas.0914710107] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Refractory temporal lobe epilepsy (TLE) is associated with a dysfunction of inhibitory signaling mediated by GABA(A) receptors. In particular, the use-dependent decrease (run-down) of the currents (I(GABA)) evoked by the repetitive activation of GABA(A) receptors is markedly enhanced in hippocampal and cortical neurons of TLE patients. Understanding the role of I(GABA) run-down in the disease, and its mechanisms, may allow development of medical alternatives to surgical resection, but such mechanistic insights are difficult to pursue in surgical human tissue. Therefore, we have used an animal model (pilocarpine-treated rats) to identify when and where the increase in I(GABA) run-down occurs in the natural history of epilepsy. We found: (i) that the increased run-down occurs in the hippocampus at the time of the first spontaneous seizure (i.e., when the diagnosis of epilepsy is made), and then extends to the neocortex and remains constant in the course of the disease; (ii) that the phenomenon is strictly correlated with the occurrence of spontaneous seizures, because it is not observed in animals that do not become epileptic. Furthermore, initial exploration of the molecular mechanism disclosed a relative increase in alpha4-, relative to alpha1-containing GABA(A) receptors, occurring at the same time when the increased run-down appears, suggesting that alterations in the molecular composition of the GABA receptors may be responsible for the occurrence of the increased run-down. These observations disclose research opportunities in the field of epileptogenesis that may lead to a better understanding of the mechanism whereby a previously normal tissue becomes epileptic.
Collapse
|
45
|
Papke RL, Stokes C. Working with OpusXpress: methods for high volume oocyte experiments. Methods 2010; 51:121-33. [PMID: 20085813 DOI: 10.1016/j.ymeth.2010.01.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 11/17/2022] Open
Abstract
OpusXpress is a semi-automated system for high throughput voltage clamp recording from Xenopus oocytes. We participated in the development process for this system and were the only laboratory to field test a prototype. Subsequently, we obtained an early production model that we have used on a regular basis for the last seven years, conducting many thousands of experiments, publishing extensively, and carrying out collaborative research in drug discovery. In this article, we relate our experience with the OpusXpress recording system and large volume oocyte handling. We provide our standard operating procedures and outline the organization of our successful team. Some of our advice is specific to researchers fortunate enough to have access to an OpusXpress system, but most of it is applicable to any group using Xenopus oocytes for the heterologous expression of ion channels.
Collapse
Affiliation(s)
- Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida, College of Medicine, Gainesville, FL 32610, USA.
| | | |
Collapse
|
46
|
Blockage of A2A and A3 adenosine receptors decreases the desensitization of human GABA(A) receptors microtransplanted to Xenopus oocytes. Proc Natl Acad Sci U S A 2009; 106:15927-31. [PMID: 19721003 DOI: 10.1073/pnas.0907324106] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We previously found that the endogenous anticonvulsant adenosine, acting through A(2A) and A(3) adenosine receptors (ARs), alters the stability of currents (I(GABA)) generated by GABA(A) receptors expressed in the epileptic human mesial temporal lobe (MTLE). Here we examined whether ARs alter the stability (desensitization) of I(GABA) expressed in focal cortical dysplasia (FCD) and in periglioma epileptic tissues. The experiments were performed with tissues from 23 patients, using voltage-clamp recordings in Xenopus oocytes microinjected with membranes isolated from human MTLE and FCD tissues or using patch-clamp recordings of pyramidal neurons in epileptic tissue slices. On repetitive activation, the epileptic GABA(A) receptors revealed instability, manifested by a large I(GABA) rundown, which in most of the oocytes (approximately 70%) was obviously impaired by the new A(2A) antagonists ANR82, ANR94, and ANR152. In most MTLE tissue-microtransplanted oocytes, a new A(3) receptor antagonist (ANR235) significantly improved I(GABA) stability. Moreover, patch-clamped pyramidal neurons from human neocortical slices of periglioma epileptic tissues exhibited altered I(GABA) rundown on ANR94 treatment. Our findings indicate that antagonizing A(2A) and A(3) receptors increases the I(GABA) stability in different epileptic tissues and suggest that adenosine derivatives may offer therapeutic opportunities in various forms of human epilepsy.
Collapse
|
47
|
Microtransplantation of ligand-gated receptor-channels from fresh or frozen nervous tissue into Xenopus oocytes: A potent tool for expanding functional information. Prog Neurobiol 2009; 88:32-40. [DOI: 10.1016/j.pneurobio.2009.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 12/21/2008] [Accepted: 01/29/2009] [Indexed: 02/05/2023]
|
48
|
Abstract
Voltage-clamp techniques are typically used to study the plasma membrane proteins, such as ion channels and transporters that control bioelectrical signals. Many of these proteins have been cloned and can now be studied as potential targets for drug development. The two approaches most commonly used for heterologous expression of cloned ion channels and transporters involve either transfection of the genes into small cells grown in tissue culture or the injection of the genetic material into larger cells. The standard large cells used for the expression of cloned cDNA or synthetic RNA are the egg progenitor cells (oocytes) of the African frog, Xenopus laevis. Until recently, cellular electrophysiology was performed manually by a single operator, one cell at a time. However, methods of high throughput electrophysiology have been developed which are automated and permit data acquisition and analysis from multiple cells in parallel. These methods are breaking a bottleneck in drug discovery, useful in some cases for primary screening as well as for thorough characterization of new drugs. Increasing throughput of high-quality functional data greatly augments the efficiency of academic research and pharmaceutical drug development. Some examples of studies that benefit most from high throughput electrophysiology include pharmaceutical screening of targeted compound libraries, secondary screening of identified compounds for subtype selectivity, screening mutants of ligand-gated channels for changes in receptor function, scanning mutagenesis of protein segments, and mutant-cycle analysis. We describe here the main features and potential applications of OpusXpress, an efficient commercially available system for automated recording from Xenopus oocytes. We show some types of data that have been gathered by this system and review realized and potential applications.
Collapse
Affiliation(s)
- Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, Florida, USA.
| | | |
Collapse
|
49
|
Roseti C, Martinello K, Fucile S, Piccari V, Mascia A, Di Gennaro G, Quarato PP, Manfredi M, Esposito V, Cantore G, Arcella A, Simonato M, Fredholm BB, Limatola C, Miledi R, Eusebi F. Adenosine receptor antagonists alter the stability of human epileptic GABAA receptors. Proc Natl Acad Sci U S A 2008; 105:15118-23. [PMID: 18809912 PMCID: PMC2567502 DOI: 10.1073/pnas.0807277105] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Indexed: 12/26/2022] Open
Abstract
We examined how the endogenous anticonvulsant adenosine might influence gamma-aminobutyric acid type A (GABA(A)) receptor stability and which adenosine receptors (ARs) were involved. Upon repetitive activation (GABA 500 microM), GABA(A) receptors, microtransplanted into Xenopus oocytes from neurosurgically resected epileptic human nervous tissues, exhibited an obvious GABA(A)-current (I(GABA)) run-down, which was consistently and significantly reduced by treatment with the nonselective adenosine receptor antagonist CGS15943 (100 nM) or with adenosine deaminase (ADA) (1 units/ml), that inactivates adenosine. It was also found that selective antagonists of A2B (MRS1706, 10 nM) or A3 (MRS1334, 30 nM) receptors reduced I(GABA) run-down, whereas treatment with the specific A1 receptor antagonist DPCPX (10 nM) was ineffective. The selective A2A receptor antagonist SCH58261 (10 nM) reduced or potentiated I(GABA) run-down in approximately 40% and approximately 20% of tested oocytes, respectively. The ADA-resistant, AR agonist 2-chloroadenosine (2-CA) (10 microM) potentiated I(GABA) run-down but only in approximately 20% of tested oocytes. CGS15943 administration again decreased I(GABA) run-down in patch-clamped neurons from either human or rat neocortex slices. I(GABA) run-down in pyramidal neurons was equivalent in A1 receptor-deficient and wt neurons but much larger in neurons from A2A receptor-deficient mice, indicating that, in mouse cortex, GABA(A)-receptor stability is tonically influenced by A2A but not by A1 receptors. I(GABA) run-down from wt mice was not affected by 2-CA, suggesting maximal ARs activity by endogenous adenosine. Our findings strongly suggest that cortical A2-A3 receptors alter the stability of GABA(A) receptors, which could offer therapeutic opportunities.
Collapse
Affiliation(s)
- Cristina Roseti
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
| | | | - Sergio Fucile
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | - Vanessa Piccari
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | | | | | | | - Mario Manfredi
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | | | | | | | - Michele Simonato
- Department of Clinical and Experimental Medicine, Section of Pharmacology, Neuroscience Center, University of Ferrara and National Institute of Neuroscience, I-44100 Ferrara, Italy
| | - Bertil B. Fredholm
- Department of Physiology and Pharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Cristina Limatola
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | - Ricardo Miledi
- **Instituto de Neurobiología, Campus UNAM-Juriquilla, Universidad Nacional Autónoma de México, AP1-1141 Querétaro, Mexico; and
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550
| | - Fabrizio Eusebi
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| |
Collapse
|
50
|
A Rosetta stone for analysis of human membrane protein function. Proc Natl Acad Sci U S A 2008; 105:10641-2. [PMID: 18669649 DOI: 10.1073/pnas.0806110105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|