1
|
Deleye L, Franchi F, Trevisani M, Loiacono F, Vercellino S, Debellis D, Liessi N, Armirotti A, Vázquez E, Valente P, Castagnola V, Benfenati F. Few-layered graphene increases the response of nociceptive neurons to irritant stimuli. NANOSCALE 2024; 16:2419-2431. [PMID: 38226500 DOI: 10.1039/d3nr03790h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The unique properties of few-layered graphene (FLG) make it interesting for a variety of applications, including biomedical applications, such as tissue engineering and drug delivery. Although different studies focus on applications in the central nervous system, its interaction with the peripheral nervous system has been so far overlooked. Here, we investigated the effects of exposure to colloidal dispersions of FLG on the sensory neurons of the rat dorsal root ganglia (DRG). We found that the FLG flakes were actively internalized by sensory neurons, accumulated in large intracellular vesicles, and possibly degraded over time, without major toxicological concerns, as neuronal viability, morphology, protein content, and basic electrical properties of DRG neurons were preserved. Interestingly, in our electrophysiological investigation under noxious stimuli, we observed an increased functional response upon FLG treatment of the nociceptive subpopulation of DRG neurons in response to irritants specific for chemoreceptors TRPV1 and TRPA1. The observed effects of FLG on DRG neurons may open-up novel opportunities for applications of these materials in specific disease models.
Collapse
Affiliation(s)
- Lieselot Deleye
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - Francesca Franchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Martina Trevisani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Fabrizio Loiacono
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Silvia Vercellino
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, IIT, Via Morego 30, 16163, Genoa, Italy
| | - Nara Liessi
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Ester Vázquez
- Facultad de Ciencias Químicas, Universidad Castilla La-Mancha, Ciudad Real, 13071 Spain
| | - Pierluigi Valente
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
2
|
Xie Z, Abumaria N. Effect of truncation on TRPM7 channel activity. Channels (Austin) 2023; 17:2200874. [PMID: 37040321 PMCID: PMC10761173 DOI: 10.1080/19336950.2023.2200874] [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: 11/20/2022] [Accepted: 04/04/2023] [Indexed: 04/12/2023] Open
Abstract
Transient receptor potential melastatin-like 7 (TRPM7) is a key player in various physiological and pathological processes. TRPM7 channel activity is regulated by different factors. The effects of cleavage of different domains on channel activity remain unknown. Here, we constructed several TRPM7 clones and explored the effects of truncating the mouse TRPM7 at different locations on the ion channel activity in two cell lines. We compared the clones' activity with the full-length TRPM7 and the native TRPM7 in transfected and untransfected cells. We also expressed fluorescently tagged truncated clones to examine their protein stability and membrane targeting. We found that truncating the kinase domain induced reduction in TRPM7 channel activity. Further truncations beyond the kinase (serine/threonine rich domain and/or coiled-coil domain) did not result in further reductions in channel activity. Two truncated clones lacking the TRP domain or the melastatin homology domain had a completely nonfunctional channel apparently due to disruption of protein stability. We identified the shortest structure of TRPM7 with measurable channel activity. We found that the truncated TRPM7 containing only S5 and S6 domains retained some channel activity. Adding the TRP domain to the S5-S6 resulted in a significant increase in channel activity. Finally, our analysis showed that TRPM7 outward currents are more sensitive to truncations than inward currents. Our data provide insights on the effects of truncating TRPM7 at different locations on the channel functions, highlighting the importance of different domains in impacting channel activity, protein stability, and/or membrane targeting.
Collapse
Affiliation(s)
- Zhuqing Xie
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Progress in the Structural Basis of thermoTRP Channel Polymodal Gating. Int J Mol Sci 2023; 24:ijms24010743. [PMID: 36614186 PMCID: PMC9821180 DOI: 10.3390/ijms24010743] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The thermosensory transient receptor potential (thermoTRP) family of ion channels is constituted by several nonselective cation channels that are activated by physical and chemical stimuli functioning as paradigmatic polymodal receptors. Gating of these ion channels is achieved through changes in temperature, osmolarity, voltage, pH, pressure, and by natural or synthetic chemical compounds that directly bind to these proteins to regulate their activity. Given that thermoTRP channels integrate diverse physical and chemical stimuli, a thorough understanding of the molecular mechanisms underlying polymodal gating has been pursued, including the interplay between stimuli and differences between family members. Despite its complexity, recent advances in cryo-electron microscopy techniques are facilitating this endeavor by providing high-resolution structures of these channels in different conformational states induced by ligand binding or temperature that, along with structure-function and molecular dynamics, are starting to shed light on the underlying allosteric gating mechanisms. Because dysfunctional thermoTRP channels play a pivotal role in human diseases such as chronic pain, unveiling the intricacies of allosteric channel gating should facilitate the development of novel drug-based resolving therapies for these disorders.
Collapse
|
4
|
Activation of TRPV1 by capsaicin-loaded CaCO3 nanoparticle for tumor-specific therapy. Biomaterials 2022; 284:121520. [DOI: 10.1016/j.biomaterials.2022.121520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/21/2022] [Accepted: 04/09/2022] [Indexed: 01/07/2023]
|
5
|
Mannes M, Martin C, Menet C, Ballet S. Wandering beyond small molecules: peptides as allosteric protein modulators. Trends Pharmacol Sci 2021; 43:406-423. [PMID: 34857409 DOI: 10.1016/j.tips.2021.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022]
Abstract
Recent years have seen the rise of allosteric modulation as an innovative approach for drug design and discovery, efforts which culminated in the development of several clinical candidates. Allosteric modulation of many drug targets, including mainly membrane-embedded receptors, have been vastly explored through small molecule screening campaigns, but much less attention has been paid to peptide-based allosteric modulators. However, peptides have a significant impact on the pharmaceutical industry due to the typically higher potency and selectivity for their targets, as compared with small molecule therapeutics. Therefore, peptides represent one of the most promising classes of molecules that can modulate key biological pathways. Here, we report on the allosteric modulation of proteins (ranging from G protein-coupled receptors to specific protein-protein interactions) by peptides for applications in drug discovery.
Collapse
Affiliation(s)
- Morgane Mannes
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium.
| | - Christel Menet
- Confo Therapeutics N.V., Technologiepark-Zwijnaarde 30, Ghent, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium.
| |
Collapse
|
6
|
Fernández-Ballester G, Fernández-Carvajal A, Ferrer-Montiel A. Targeting thermoTRP ion channels: in silico preclinical approaches and opportunities. Expert Opin Ther Targets 2020; 24:1079-1097. [PMID: 32972264 DOI: 10.1080/14728222.2020.1820987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION A myriad of cellular pathophysiological responses are mediated by polymodal ion channels that respond to chemical and physical stimuli such as thermoTRP channels. Intriguingly, these channels are pivotal therapeutic targets with limited clinical pharmacology. In silico methods offer an unprecedented opportunity for discovering new lead compounds targeting thermoTRP channels with improved pharmacological activity and therapeutic index. AREAS COVERED This article reviews the progress on thermoTRP channel pharmacology because of (i) advances in solving their atomic structure using cryo-electron microscopy and, (ii) progress on computational techniques including homology modeling, molecular docking, virtual screening, molecular dynamics, ADME/Tox and artificial intelligence. Together, they have increased the number of lead compounds with clinical potential to treat a variety of pathologies. We used original and review articles from Pubmed (1997-2020), as well as the clinicaltrials.gov database, containing the terms thermoTRP, artificial intelligence, docking, and molecular dynamics. EXPERT OPINION The atomic structure of thermoTRP channels along with computational methods constitute a realistic first line strategy for designing drug candidates with improved pharmacology and clinical translation. In silico approaches can also help predict potential side-effects that can limit clinical development of drug candidates. Together, they should provide drug candidates with upgraded therapeutic properties.
Collapse
Affiliation(s)
- Gregorio Fernández-Ballester
- Professor Gregorio Fernández-Ballester. Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández , Alicante, Spain
| | - Asia Fernández-Carvajal
- Professor Gregorio Fernández-Ballester. Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández , Alicante, Spain
| | - Antonio Ferrer-Montiel
- Professor Gregorio Fernández-Ballester. Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández , Alicante, Spain
| |
Collapse
|
7
|
Escelsior A, Sterlini B, Murri MB, Serafini G, Aguglia A, da Silva BP, Corradi A, Valente P, Amore M. Red-hot chili receptors: A systematic review of TRPV1 antagonism in animal models of psychiatric disorders and addiction. Behav Brain Res 2020; 393:112734. [DOI: 10.1016/j.bbr.2020.112734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022]
|
8
|
Xian W, Wang H, Moretti A, Laugwitz KL, Flockerzi V, Lipp P. Domain zipping and unzipping modulates TRPM4's properties in human cardiac conduction disease. FASEB J 2020; 34:12114-12126. [PMID: 32681584 DOI: 10.1096/fj.202000097rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 11/11/2022]
Abstract
The transient receptor potential melastatin 4 (TRPM4) is a Ca2+ -activated nonselective cation channel linked to human cardiac diseases. The human mutation K914R within TRPM4's S4-S5 linker was identified in patients with atrioventricular block. During UV-flash-mediated Ca2+ transients, TRPM4K914R generated a threefold augmented membrane current concomitant with 2 to 3-fold slowed down activation and deactivation kinetics resulting in excessive membrane currents during human cardiac action potentials. Mutagenesis of K914 paired with molecular modeling suggested the importance of the nanoscopic interface between the S4-S5 linker, the MHR4-, and TRP-domain as a major determinant for TRPM4's behavior. Rational mutagenesis of an interacting amino acid (R1062Q) in the TRP domain was able to offset K914R`s gain-of-function by zipping and unzipping of this nanoscopic interface. In conclusion, repulsion and attraction between the amino acids at positions 914 and 1062 alters the flexibility of the nanoscopic interface suggesting a zipping and unzipping mechanism that modulates TRPM4's functions. Pharmacological modulation of this intramolecular mechanism might represent a novel therapeutic strategy for the management of TRPM4-mediated cardiac diseases.
Collapse
Affiliation(s)
- Wenying Xian
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, Homburg, Germany
| | - Hongmei Wang
- Experimental and Clinical Pharmacology and Toxicology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, Homburg, Germany
| | - Alessandra Moretti
- First Medical Department (Cardiology), Klinikum rechts der Isar, Technische Universität München, München, Germany.,DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- First Medical Department (Cardiology), Klinikum rechts der Isar, Technische Universität München, München, Germany.,DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, Homburg, Germany
| | - Peter Lipp
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, Homburg, Germany
| |
Collapse
|
9
|
Zheng W, Cai R, Hofmann L, Nesin V, Hu Q, Long W, Fatehi M, Liu X, Hussein S, Kong T, Li J, Light PE, Tang J, Flockerzi V, Tsiokas L, Chen XZ. Direct Binding between Pre-S1 and TRP-like Domains in TRPP Channels Mediates Gating and Functional Regulation by PIP2. Cell Rep 2019; 22:1560-1573. [PMID: 29425510 PMCID: PMC6483072 DOI: 10.1016/j.celrep.2018.01.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/08/2017] [Accepted: 01/11/2018] [Indexed: 11/28/2022] Open
Abstract
Transient receptor potential (TRP) channels are regulated by diverse stimuli comprising thermal, chemical, and mechanical modalities. They are also commonly regulated by phosphatidylinositol-4,5-bisphosphate (PIP2), with underlying mechanisms largely unknown. We here revealed an intramolecular interaction of the TRPP3 N and C termini (N-C) that is functionally essential. The interaction was mediated by aromatic Trp81 in pre-S1 domain and cationic Lys568 in TRP-like domain. Structure-function analyses revealed similar N-C interaction in TRPP2 as well as TRPM8/-V1/-C4 via highly conserved tryptophan and lysine/arginine residues. PIP2 bound to cationic residues in TRPP3, including K568, thereby disrupting the N-C interaction and negatively regulating TRPP3. PIP2 had similar negative effects on TRPP2. Interestingly, we found that PIP2 facilitates the N-C interaction in TRPM8/-V1, resulting in channel potentiation. The intramolecular N-C interaction might represent a shared mechanism underlying the gating and PIP2 regulation of TRP channels. Zheng et al. show that an aromatic Trp residue in pre-S1 and a cationic Lys residue in the TRP-like domain of TRP polycystin channels mediate N-C binding, which underlies TRPPs gating and PIP2 regulation. The conservation of these residues suggests that this may be a shared mechanism of TRP channel gating.
Collapse
Affiliation(s)
- Wang Zheng
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei 430068, China; Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ruiqi Cai
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg 66421, Germany
| | - Vasyl Nesin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Qiaolin Hu
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Wentong Long
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Mohammad Fatehi
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Xiong Liu
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Shaimaa Hussein
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Tim Kong
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Jingru Li
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Peter E Light
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei 430068, China.
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg 66421, Germany
| | - Leonidas Tsiokas
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Xing-Zhen Chen
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei 430068, China; Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
10
|
Abstract
The implication of several TRP ion channels (e.g., TRPV1) in diverse physiological and pathological processes has signaled them as pivotal drug targets. Consequently, the identification of selective and potent ligands for these channels is of great interest in pharmacology and biomedicine. However, a major challenge in the design of modulators is ensuring the specificity for their intended targets. In recent years, the emergence of high-resolution structures of ion channels facilitates the computer-assisted drug design at molecular levels. Here we describe some computational methods and general protocols to discover channel modulators, including homology modelling, docking and virtual screening, and structure-based peptide design.
Collapse
Affiliation(s)
- Magdalena Nikolaeva Koleva
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universitas Miguel Hernández, Elche, Spain
- AntalGenics SL. Ed. Quorum III, University Scientific Park, Universitas Miguel Hernández, Elche, Spain
| | - Gregorio Fernandez-Ballester
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universitas Miguel Hernández, Elche, Spain.
| |
Collapse
|
11
|
Pérez de Vega MJ, Ferrer-Montiel A, González-Muñiz R. Recent progress in non-opioid analgesic peptides. Arch Biochem Biophys 2018; 660:36-52. [DOI: 10.1016/j.abb.2018.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 02/08/2023]
|
12
|
Abstract
The sensation of pain plays a vital protecting role, alerting organisms about potentially damaging stimuli. Tissue injury is detected by nerve endings of specialized peripheral sensory neurons called nociceptors that are equipped with different ion channels activated by thermal, mechanic, and chemical stimuli. Several transient receptor potential channels have been identified as molecular transducers of thermal stimuli in pain-sensing neurons. Skin injury or inflammation leads to increased sensitivity to thermal and mechanic stimuli, clinically defined as allodynia or hyperalgesia. This hypersensitivity is also characteristic of systemic inflammatory disorders and neuropathic pain conditions. Mechanisms of thermal hyperalgesia include peripheral sensitization of nociceptor afferents and maladaptive changes in pain-encoding neurons within the central nervous system. An important aspect of pain management involves attempts to minimize the development of nociceptor hypersensitivity. However, knowledge about the cellular and molecular mechanisms causing thermal hyperalgesia and allodynia in human subjects is still limited, and such knowledge would be an essential step for the development of more effective therapies.
Collapse
Affiliation(s)
- Félix Viana
- Alicante Institute of Neurosciences, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Spain.
| |
Collapse
|
13
|
Heymann HM, Wu Y, Lu Y, Qvit N, Gross GJ, Gross ER. Transient receptor potential vanilloid 1 inhibitors block laparotomy- and opioid-induced infarct size reduction in rats. Br J Pharmacol 2017; 174:4826-4835. [PMID: 28982207 DOI: 10.1111/bph.14064] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/27/2017] [Accepted: 09/28/2017] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE In light of the opioid epidemic, physicians are increasingly prescribing non-opioid analgesics to surgical patients. Transient receptor potential vanilloid 1 (TRPV1) inhibitors are potentially alternative pain therapeutics for surgery. Here, we examined in rodents whether the cardioprotection conferred by two common procedures during surgery, a laparotomy or morphine delivery, is mediated by the TRPV1 channel. We further tested whether an experimental analgesic peptide (known as P5) targeted against the TRPV1 C-terminus region interferes with laparotomy- or morphine-induced cardioprotection. EXPERIMENTAL APPROACH Male Sprague-Dawley rats were subjected to 30 min coronary occlusion followed by 120 min reperfusion. Before ischaemia, a laparotomy with or without capsaicin application (0.1% cream, a TRPV1 activator) was performed. Additional rats were given morphine (0.3 mg·kg-1 ) with or without capsaicin. In addition, capsazepine (3 mg·kg-1 , a classical TRPV1 inhibitor), or P5 (3 mg·kg-1 , a peptide analgesic and TRPV1 inhibitor), was given either alone or prior to a laparotomy or morphine administration. Myocardial infarct size was determined. KEY RESULTS A laparotomy, in addition to combining a laparotomy with capsaicin cream, reduced infarct size versus control. Morphine, in addition to combining morphine administration with capsaicin cream, also reduced infarct size versus control. When TRPV1 inhibitors capsazepine or P5 were given, either TRPV1 inhibitor abolished the infarct size reduction mediated by a laparotomy or morphine. CONCLUSIONS AND IMPLICATIONS Inhibiting the TRPV1 channel blocks laparotomy- or morphine-induced cardioprotection. Impaired organ protection may be a potential pitfall of using TRPV1 inhibitors for pain control.
Collapse
Affiliation(s)
- Helen M Heymann
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yun Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yao Lu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Nir Qvit
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Garrett J Gross
- Department of Pharmacology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
14
|
Pérez de Vega MJ, Gómez-Monterrey I, Ferrer-Montiel A, González-Muñiz R. Transient Receptor Potential Melastatin 8 Channel (TRPM8) Modulation: Cool Entryway for Treating Pain and Cancer. J Med Chem 2016; 59:10006-10029. [PMID: 27437828 DOI: 10.1021/acs.jmedchem.6b00305] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
TRPM8 ion channels, the primary cold sensors in humans, are activated by innocuous cooling (<28 °C) and cooling compounds (menthol, icilin) and are implicated in sensing unpleasant cold stimuli as well as in mammalian thermoregulation. Overexpression of these thermoregulators in prostate cancer and in other life-threatening tumors, along with their contribution to an increasing number of pathological conditions, opens a plethora of medicinal chemistry opportunities to develop receptor modulators. This Perspective seeks to describe current known modulators for this ion channel because both agonists and antagonists may be useful for the treatment of most TRPM8-mediated pathologies. We primarily focus on SAR data for the different families of compounds and the pharmacological properties of the most promising ligands. Furthermore, we also address the knowledge about the channel structure, although still in its infancy, and the role of the TRPM8 protein signalplex to channel function and dysfunction. We finally outline the potential future prospects of the challenging TRPM8 drug discovery field.
Collapse
Affiliation(s)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università "Federico II" de Napoli , Via D. Montesano 49, 80131, Naples, Italy
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular. Universitas Miguel Hernández . 03202 Alicante, Spain
| | | |
Collapse
|
15
|
Gregorio-Teruel L, Valente P, Liu B, Fernández-Ballester G, Qin F, Ferrer-Montiel A. The Integrity of the TRP Domain Is Pivotal for Correct TRPV1 Channel Gating. Biophys J 2016; 109:529-41. [PMID: 26244735 DOI: 10.1016/j.bpj.2015.06.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 01/19/2023] Open
Abstract
Transient receptor potential vanilloid subtype I (TRPV1) is a thermosensory ion channel that is also gated by chemical substances such as vanilloids. Adjacent to the channel gate, this polymodal thermoTRP channel displays a TRP domain, referred to as AD1, that plays a role in subunit association and channel gating. Previous studies have shown that swapping the AD1 in TRPV1 with the cognate from the TRPV2 channel (AD2) reduces protein expression and produces a nonfunctional chimeric channel (TRPV1-AD2). Here, we used a stepwise, sequential, cumulative site-directed mutagenesis approach, based on rebuilding the AD1 domain in the TRPV1-AD2 chimera, to unveil the minimum number of amino acids needed to restore protein expression and polymodal channel activity. Unexpectedly, we found that virtually full restitution of the AD1 sequence is required to reinstate channel expression and responses to capsaicin, temperature, and voltage. This strategy identified E692, R701, and T704 in the TRP domain as important for TRPV1 activity. Even conservative mutagenesis at these sites (E692D/R701K/T704S) impaired channel expression and abolished TRPV1 activity. However, the sole mutation of these positions in the TRPV1-AD2 chimera (D692E/K701R/S704T) was not sufficient to rescue channel gating, implying that other residues in the TRP domain are necessary to endow activity to TRPV1-AD2. A biophysical analysis of a functional chimera suggested that mutations in the TRP domain raised the energetics of channel gating by altering the coupling of stimuli sensing and pore opening. These findings indicate that inter- and/or intrasubunit interactions in the TRP domain are essential for correct TRPV1 gating.
Collapse
Affiliation(s)
- Lucia Gregorio-Teruel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain
| | - Pierluigi Valente
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genova, Italy
| | - Beiying Liu
- Department of Physiology and Biophysical Sciences, State University of New York, Buffalo, New York
| | | | - Feng Qin
- Department of Physiology and Biophysical Sciences, State University of New York, Buffalo, New York
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain.
| |
Collapse
|
16
|
Lee YS, Park JS, Jung SM, Kim SD, Kim JH, Lee JY, Jung KC, Mamura M, Lee S, Kim SJ, Bae YS, Park SH. Inhibition of lethal inflammatory responses through the targeting of membrane-associated Toll-like receptor 4 signaling complexes with a Smad6-derived peptide. EMBO Mol Med 2016; 7:577-92. [PMID: 25766838 PMCID: PMC4492818 DOI: 10.15252/emmm.201404653] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We have previously reported that Smad6, one of the inhibitory Smads of transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signaling, inhibits Toll-like receptor (TLR) 4 signaling by disrupting the Pellino-1-mediated TLR4 signaling complex. Here, we developed Smaducin-6, a novel membrane-tethered palmitic acid-conjugated Smad6-derived peptide composed of amino acids 422–441 of Smad6. Smaducin-6 interacted with Pellino-1, located in the inner membrane, thereby disrupting the formation of IRAK1-, RIP1-, IKKε-mediated TLR4 signaling complexes. Systemic administration of Smaducin-6 showed a significant therapeutic effect on mouse TLR4-mediated inflammatory disease models, cecal-ligation–puncture (CLP)-induced sepsis, and lipopolysaccharide-induced endotoxemia, by inhibiting pro-inflammatory cytokine production and apoptosis while enhancing neutrophil migration and bacterial clearance. Our findings provide clues to develop new peptide-based drugs to target Pellino-1 protein in TLR4 signaling pathway for the treatment of sepsis.
Collapse
Affiliation(s)
- Youn Sook Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jin Seok Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Sang-Doo Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jun Hwan Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jae Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Kyeong Cheon Jung
- Department of Pathology, College of Medicine, Seoul National University, Seoul, Korea
| | - Mizuko Mamura
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | | | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
17
|
Calcium Entry Through Thermosensory Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:265-304. [PMID: 27161233 DOI: 10.1007/978-3-319-26974-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic β-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.
Collapse
|
18
|
Valente P, Orlando M, Raimondi A, Benfenati F, Baldelli P. Fine Tuning of Synaptic Plasticity and Filtering by GABA Released from Hippocampal Autaptic Granule Cells. Cereb Cortex 2015; 26:1149-67. [PMID: 25576534 DOI: 10.1093/cercor/bhu301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The functional consequence of γ-aminobutyric acid (GABA) release at mossy fiber terminals is still a debated topic. Here, we provide multiple evidence of GABA release in cultured autaptic hippocampal granule cells. In ∼50% of the excitatory autaptic neurons, GABA, VGAT, or GAD67 colocalized with vesicular glutamate transporter 1-positive puncta, where both GABAB and GABAA receptors (Rs) were present. Patch-clamp recordings showed a clear enhancement of autaptic excitatory postsynaptic currents in response to the application of the GABABR antagonist CGP58845 only in neurons positive to the selective granule cell marker Prox1, and expressing low levels of GAD67. Indeed, GCP non-responsive excitatory autaptic neurons were both Prox1- and GAD67-negative. Although the amount of released GABA was not sufficient to activate functional postsynaptic GABAARs, it effectively activated presynaptic GABABRs that maintain a tonic "brake" on the probability of release and on the size of the readily releasable pool and contributed to resting potential hyperpolarization possibly through extrasynaptic GABAAR activation. The autocrine inhibition exerted by GABABRs on glutamate release enhanced both paired-pulse facilitation and post-tetanic potentiation. Such GABABR-mediated changes in short-term plasticity confer to immature granule cells the capability to modulate their filtering properties in an activity-dependent fashion, with remarkable consequences on the dynamic behavior of neural circuits.
Collapse
Affiliation(s)
- Pierluigi Valente
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy Department of Experimental Medicine, Section of Physiology, University of Genova, Genova 16132, Italy
| | - Marta Orlando
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Andrea Raimondi
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy Department of Experimental Medicine, Section of Physiology, University of Genova, Genova 16132, Italy
| | - Pietro Baldelli
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy Department of Experimental Medicine, Section of Physiology, University of Genova, Genova 16132, Italy
| |
Collapse
|
19
|
Abstract
Lipopeptides based on the intracellular loops of cell-surface receptors, known as "Pepducins," represent a promising new class of compounds used for the study of membrane proteins and as potential therapeutics in a variety of diseases. Detailed knowledge of the three-dimensional structure of G-protein-coupled receptors (GPCRs) and delineation of the mechanisms of pepducin activation and biased G-protein signaling has facilitated the development of even more potent pepducin allosteric modulators.
Collapse
|
20
|
Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, Sessa A, Colasante G, Bartolomeo R, Massimino L, Ferroni S, Settembre C, Benfenati F, Broccoli V. Direct conversion of fibroblasts into functional astrocytes by defined transcription factors. Stem Cell Reports 2014; 4:25-36. [PMID: 25556566 PMCID: PMC4297873 DOI: 10.1016/j.stemcr.2014.12.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/04/2023] Open
Abstract
Direct cell reprogramming enables direct conversion of fibroblasts into functional neurons and oligodendrocytes using a minimal set of cell-lineage-specific transcription factors. This approach is rapid and simple, generating the cell types of interest in one step. However, it remains unknown whether this technology can be applied to convert fibroblasts into astrocytes, the third neural lineage. Astrocytes play crucial roles in neuronal homeostasis, and their dysfunctions contribute to the origin and progression of multiple human diseases. Herein, we carried out a screening using several transcription factors involved in defining the astroglial cell fate and identified NFIA, NFIB, and SOX9 to be sufficient to convert with high efficiency embryonic and postnatal mouse fibroblasts into astrocytes (iAstrocytes). We proved both by gene-expression profiling and functional tests that iAstrocytes are comparable to native brain astrocytes. This protocol can be then employed to generate functional iAstrocytes for a wide range of experimental applications. NFIA, NFIB, and SOX9 reprogram fibroblasts into induced astrocytes (iAstrocytes) iAstrocytes reprogramming induces a global change in gene-expression profiling iAstrocytes are functionally comparable to native astrocytes NFIA, NFIB, and SOX9 induce an astrocytic phenotype in human fibroblasts
Collapse
Affiliation(s)
- Massimiliano Caiazzo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| | - Serena Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Pierluigi Valente
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy
| | - Gabriele Lignani
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | | | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rosa Bartolomeo
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Medical Genetics, Department of Medical and Translational Science Unit, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Fabio Benfenati
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy; Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| |
Collapse
|
21
|
Gregorio-Teruel L, Valente P, González-Ros JM, Fernández-Ballester G, Ferrer-Montiel A. Mutation of I696 and W697 in the TRP box of vanilloid receptor subtype I modulates allosteric channel activation. ACTA ACUST UNITED AC 2014; 143:361-75. [PMID: 24567510 PMCID: PMC3933934 DOI: 10.1085/jgp.201311070] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Residues I696 and W697 are crucial to coupling between the TRPV1 ligand- and voltage-sensing domains and the channel pore. The transient receptor potential vanilloid receptor subtype I (TRPV1) channel acts as a polymodal sensory receptor gated by chemical and physical stimuli. Like other TRP channels, TRPV1 contains in its C terminus a short, conserved domain called the TRP box, which is necessary for channel gating. Substitution of two TRP box residues—I696 and W697—with Ala markedly affects TRPV1’s response to all activating stimuli, which indicates that these two residues play a crucial role in channel gating. We systematically replaced I696 and W697 with 18 native l-amino acids (excluding cysteine) and evaluated the effect on voltage- and capsaicin-dependent gating. Mutation of I696 decreased channel activation by either voltage or capsaicin; furthermore, gating was only observed with substitution of hydrophobic amino acids. Substitution of W697 with any of the 18 amino acids abolished gating in response to depolarization alone, shifting the threshold to unreachable voltages, but not capsaicin-mediated gating. Moreover, vanilloid-activated responses of W697X mutants showed voltage-dependent gating along with a strong voltage-independent component. Analysis of the data using an allosteric model of activation indicates that mutation of I696 and W697 primarily affects the allosteric coupling constants of the ligand and voltage sensors to the channel pore. Together, our findings substantiate the notion that inter- and/or intrasubunit interactions at the level of the TRP box are critical for efficient coupling of stimulus sensing and gate opening. Perturbation of these interactions markedly reduces the efficacy and potency of the activating stimuli. Furthermore, our results identify these interactions as potential sites for pharmacological intervention.
Collapse
Affiliation(s)
- Lucia Gregorio-Teruel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, 03202 Elche, Spain
| | | | | | | | | |
Collapse
|
22
|
Veldhuis NA, Poole DP, Grace M, McIntyre P, Bunnett NW. The G Protein–Coupled Receptor–Transient Receptor Potential Channel Axis: Molecular Insights for Targeting Disorders of Sensation and Inflammation. Pharmacol Rev 2014; 67:36-73. [DOI: 10.1124/pr.114.009555] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
23
|
The design and implementation of a generic lipopeptide scanning platform to enable the identification of ‘locally acting’ agonists for the apelin receptor. Bioorg Med Chem Lett 2014; 24:4871-5. [DOI: 10.1016/j.bmcl.2014.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/15/2014] [Accepted: 08/20/2014] [Indexed: 12/19/2022]
|
24
|
Taberner FJ, López-Córdoba A, Fernández-Ballester G, Korchev Y, Ferrer-Montiel A. The region adjacent to the C-end of the inner gate in transient receptor potential melastatin 8 (TRPM8) channels plays a central role in allosteric channel activation. J Biol Chem 2014; 289:28579-94. [PMID: 25157108 DOI: 10.1074/jbc.m114.577478] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ability of transient receptor potential (TRP) channels to sense and respond to environmental and endogenous cues is crucial in animal sensory physiology. The molecular mechanism of channel gating is yet elusive. The TRP box, a conserved region in the N-end of the C terminus domain, has been signaled as pivotal for allosteric activation in TRP channels. Here, we have examined the role of the linker region between the TRPM8 inner gate and the TRP box (referred to as the S6-TRP box linker) to identify structural determinants of channel gating. Stepwise substitutions of segments in the S6-TRP box linker of TRPM8 channel with the cognate TRPV1 channel sequences produced functional chimeric channels, and identified Tyr(981) as a central molecular determinant of channel function. Additionally, mutations in the 986-990 region had a profound impact on channel gating by voltage and menthol, as evidenced by the modulation of the conductance-to-voltage (G-V) relationships. Simulation of G-V curves using an allosteric model for channel activation revealed that these mutations altered the allosteric constants that couple stimuli sensing to pore opening. A molecular model of TRPM8, based on the recently reported TRPV1 structural model, showed that Tyr(981) may lie in a hydrophobic pocket at the end of the S6 transmembrane segment and is involved in inter-subunit interactions with residues from neighbor subunits. The 986-990 region holds intrasubunit interactions between the TRP domain and the S4-S5 linker. These findings substantiate a gating mechanism whereby the TRP domain acts as a coupling domain for efficient channel opening. Furthermore, they imply that protein-protein interactions of the TRP domain may be targets for channel modulation and drug intervention.
Collapse
Affiliation(s)
- Francisco José Taberner
- From the Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Ainara López-Córdoba
- From the Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, 03202 Elche, Spain
| | | | - Yuri Korchev
- the Imperial College School of Medicine, SW7 2AZ London, United Kingdom, and
| | - Antonio Ferrer-Montiel
- From the Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, 03202 Elche, Spain, the Unidad de Biofísica, UPV/EHU, CSIC, 48940 Leioa, Spain
| |
Collapse
|
25
|
Bonache MÁ, Balsera B, López-Méndez B, Millet O, Brancaccio D, Gómez-Monterrey I, Carotenuto A, Pavone LM, Reille-Seroussi M, Gagey-Eilstein N, Vidal M, de la Torre-Martı́nez R, Fernández-Carvajal A, Ferrer-Montiel A, García-López MT, Martín-Martínez M, de Vega MJP, González-Muñiz R. De novo designed library of linear helical peptides: an exploratory tool in the discovery of protein-protein interaction modulators. ACS COMBINATORIAL SCIENCE 2014; 16:250-8. [PMID: 24725184 DOI: 10.1021/co500005x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-protein interactions (PPIs) have emerged as important targets for pharmaceutical intervention because of their essential role in numerous physiological and pathological processes, but screening efforts using small-molecules have led to very low hit rates. Linear peptides could represent a quick and effective approach to discover initial PPI hits, particularly if they have inherent ability to adopt specific peptide secondary structures. Here, we address this hypothesis through a linear helical peptide library, composed of four sublibraries, which was designed by theoretical predictions of helicity (Agadir software). The 13-mer peptides of this collection fixes either a combination of three aromatic or two aromatic and one aliphatic residues on one face of the helix (Ac-SSEEX(5)ARNX(9)AAX(12)N-NH2), since these are structural features quite common at PPIs interfaces. The 81 designed peptides were conveniently synthesized by parallel solid-phase methodologies, and the tendency of some representative library components to adopt the intended secondary structure was corroborated through CD and NMR experiments. As proof of concept in the search for PPI modulators, the usefulness of this library was verified on the widely studied p53-MDM2 interaction and on the communication between VEGF and its receptor Flt-1, two PPIs for which a hydrophobic α-helix is essential for the interaction. We have demonstrated here that, in both cases, selected peptides from the library, containing the right hydrophobic sequence of the hot-spot in one of the protein partners, are able to interact with the complementary protein. Moreover, we have discover some new, quite potent inhibitors of the VEGF-Flt-1 interaction, just by replacing one of the aromatic residues of the initial F(5)Y(9)Y(12) peptide by W, in agreement with previous results on related antiangiogenic peptides. Finally, the HTS evaluation of the full collection on thermoTRPs has led to a few antagonists of TRPV1 and TRPA1 channels, which open new avenues on the way to innovative modulators of these channels.
Collapse
Affiliation(s)
- M. Ángeles Bonache
- Instituto de Química-Médica (IQM-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
| | - Beatriz Balsera
- Instituto de Química-Médica (IQM-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
| | | | - Oscar Millet
- CICbioGUNE, Structural Biology Unit, 48160 Bilbao, Spain
| | - Diego Brancaccio
- Department
of Pharmacy, University of Naples “Federico II”, Via D. Montesano
49, 80131 Naples, Italy
| | - Isabel Gómez-Monterrey
- Department
of Pharmacy, University of Naples “Federico II”, Via D. Montesano
49, 80131 Naples, Italy
| | - Alfonso Carotenuto
- Department
of Pharmacy, University of Naples “Federico II”, Via D. Montesano
49, 80131 Naples, Italy
| | - Luigi M. Pavone
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131, Naples, Italy
| | - Marie Reille-Seroussi
- UMR
8638
CNRS, UFR de Pharmacie, Université Paris Descartes, PRES
Sorbonne Paris Cité, 4 avenue de l’Observatoire, 75006 Paris, France
| | - Nathalie Gagey-Eilstein
- UMR
8638
CNRS, UFR de Pharmacie, Université Paris Descartes, PRES
Sorbonne Paris Cité, 4 avenue de l’Observatoire, 75006 Paris, France
| | - Michel Vidal
- UMR
8638
CNRS, UFR de Pharmacie, Université Paris Descartes, PRES
Sorbonne Paris Cité, 4 avenue de l’Observatoire, 75006 Paris, France
- UF
“Pharmacocinétique et pharmacochimie”, Hôpital Cochin, , AP-HP, 27 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Roberto de la Torre-Martı́nez
- Instituto
de Biología Molecular y Celular, Universidad Miguel Hernández, Avenida de la Universidad s/n, 03202 Elche (Alicante), Spain
| | - Asia Fernández-Carvajal
- Instituto
de Biología Molecular y Celular, Universidad Miguel Hernández, Avenida de la Universidad s/n, 03202 Elche (Alicante), Spain
| | - Antonio Ferrer-Montiel
- Instituto
de Biología Molecular y Celular, Universidad Miguel Hernández, Avenida de la Universidad s/n, 03202 Elche (Alicante), Spain
| | | | | | | | | |
Collapse
|
26
|
Flynn R, Chapman K, Iftinca M, Aboushousha R, Varela D, Altier C. Targeting the transient receptor potential vanilloid type 1 (TRPV1) assembly domain attenuates inflammation-induced hypersensitivity. J Biol Chem 2014; 289:16675-87. [PMID: 24808184 DOI: 10.1074/jbc.m114.558668] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The transient receptor potential channel vanilloid type 1 (TRPV1) is a non-selective cation channel expressed in sensory neurons of the dorsal root and trigeminal ganglia. TRPV1 is a polymodal channel activated by noxious heat, capsaicin, and protons. As a sensor for noxious stimuli, TRPV1 channel has been described as a key contributor to pain signaling. To form a functional channel, TRPV1 subunits must assemble into tetramers, and several studies have identified the TRPV1 C terminus as an essential element in subunit association. Here we combined biochemical assays with electrophysiology and imaging-based bimolecular fluorescence complementation (BiFC) and bioluminescence resonance energy transfer (BRET) in live cells to identify a short motif in the C-terminal tail of the TRPV1 subunit that governs channel assembly. Removing this region through early truncation or targeted deletion results in loss of subunit association and channel function. Importantly, we found that interfering with TRPV1 subunit association using a plasma membrane-tethered peptide attenuated mechanical and thermal hypersensitivity in two mouse models of inflammatory hyperalgesia. This represents a novel mechanism to disrupt TRPV1 subunit assembly and hence may offer a new analgesic tool for pain relief.
Collapse
Affiliation(s)
- Robyn Flynn
- From the Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada and
| | - Kevin Chapman
- From the Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada and
| | - Mircea Iftinca
- From the Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada and
| | - Reem Aboushousha
- From the Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada and
| | - Diego Varela
- Centro de Estudios Moleculares de la Celula and Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
| | - Christophe Altier
- From the Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada and
| |
Collapse
|
27
|
Ferrer-Montiel A, Fernández-Carvajal A, Planells-Cases R, Fernández-Ballester G, González-Ros JM, Messeguer A, González-Muñiz R. Advances in modulating thermosensory TRP channels. Expert Opin Ther Pat 2012; 22:999-1017. [PMID: 22835143 DOI: 10.1517/13543776.2012.711320] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Thermosensory channels are a subfamily of the transient receptor potential (TRP) channel family that are activated by changes in the environmental temperature. These channels, known as thermoTRPs, cover the entire spectrum of temperatures, from noxious cold (< 15°C) to injurious heat (> 42°C). In addition, dysfunction of these channels contributes to the thermal hypersensitivity that accompanies painful conditions. Moreover, because of their wide tissue and cellular distribution, thermoTRPs are also involved in the pathophysiology of several diseases, from inflammation to cancer. AREAS COVERED Although the number of thermoTRPs is increasing with the identification of novel members such as TRPM3, we will cover the recent advances in the pharmacology of the classical thermosensory channels, namely TRPV1, TRPV2, TRPV3, TRPV4, TRPM8 and TRPA1. This review will focus on the therapeutic progress carried out for all these channels and will highlight the tenet that TRPV1, TRPM8 and TRPA1 are the most exploited channels, and that the interest on TRPV3 and TRPV4 is growing with the first TRPV3 antagonist that moves into Phase-II clinical trials. In contrast, the pharmacology of TRPV2 is yet in its infancy. EXPERT OPINION Despite the tremendous academic and industrial investment to develop therapeutic modulators of thermoTRPs, it apparently seems that we are still far from the first successful product, although hope is maintained high for all compounds currently in clinical trials. A major concern has been the appearance of side effects. A better knowledge of the thermosensory protein networks (signal-plexes), along with the application of system biology approaches may provide novel strategies to modulate thermoTRPs activity with improved therapeutic index. A case in point is TRPV1, where acting on interacting proteins is providing new therapeutic opportunities.
Collapse
Affiliation(s)
- Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universitas Miguel Hernández, Alicante, Spain.
| | | | | | | | | | | | | |
Collapse
|
28
|
Cao X, Yang F, Zheng J, Wang K. Intracellular proton-mediated activation of TRPV3 channels accounts for the exfoliation effect of α-hydroxyl acids on keratinocytes. J Biol Chem 2012; 287:25905-16. [PMID: 22679014 DOI: 10.1074/jbc.m112.364869] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
α-Hydroxyl acids (AHAs) from natural sources act as proton donors and topical compounds that penetrate skin and are well known in the cosmetic industry for their use in chemical peels and improvement of the skin. However, little is known about how AHAs cause exfoliation to expose fresh skin cells. Here we report that the transient receptor potential vanilloid 3 (TRPV3) channel in keratinocytes is potently activated by intracellular acidification induced by glycolic acid. Patch clamp recordings and cell death assay of both human keratinocyte HaCaT cells and TRPV3-expressing HEK-293 cells confirmed that intracellular acidification led to direct activation of TRPV3 and promoted cell death. Site-directed mutagenesis revealed that an N-terminal histidine residue, His-426, known to be involved in 2-aminoethyl diphenylborinate-mediated TRPV3 activation, is critical for sensing intracellular proton levels. Taken together, our findings suggest that intracellular protons can strongly activate TRPV3, and TRPV3-mediated proton sensing and cell death in keratinocytes may serve as a molecular basis for the cosmetic use of AHAs and their therapeutic potential in acidic pH-related skin disorders.
Collapse
Affiliation(s)
- Xu Cao
- Department of Neurobiology, Neuroscience Research Institute, Peking University Health Science Center, China
| | | | | | | |
Collapse
|
29
|
O'Callaghan K, Kuliopulos A, Covic L. Turning receptors on and off with intracellular pepducins: new insights into G-protein-coupled receptor drug development. J Biol Chem 2012; 287:12787-96. [PMID: 22374997 DOI: 10.1074/jbc.r112.355461] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are a large family of remarkably versatile membrane proteins that are attractive therapeutic targets because of their involvement in a vast range of normal physiological processes and pathological diseases. Upon activation, intracellular domains of GPCRs mediate signaling to G-proteins, but these domains have yet to be effectively exploited as drug targets. Cell-penetrating lipidated peptides called pepducins target specific intracellular loops of GPCRs and have recently emerged as effective allosteric modulators of GPCR activity. The lipid moiety facilitates translocation across the plasma membrane, where pepducins then specifically modulate signaling of their cognate receptor. To date, pepducins and related lipopeptides have been shown to specifically modulate the activity of diverse GPCRs and other membrane proteins, including protease-activated receptors (PAR1, PAR2, and PAR4), chemokine receptors (CXCR1, CXCR2, and CXCR4), sphingosine 1-phosphate receptor-3 (S1P3), the melanocortin-4 receptor, the Smoothened receptor, formyl peptide receptor-2 (FPR2), the relaxin receptor (LGR7), G-proteins (Gα(q/11/o/13)), muscarinic acetylcholine receptor and vanilloid (TRPV1) channels, and the GPIIb integrin. This minireview describes recent advances made using pepducin technology in targeting diverse GPCRs and the use of pepducins in identifying potential novel drug targets.
Collapse
Affiliation(s)
- Katie O'Callaghan
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
30
|
New strategies to develop novel pain therapies: addressing thermoreceptors from different points of view. Pharmaceuticals (Basel) 2011; 5:16-48. [PMID: 24288041 PMCID: PMC3763626 DOI: 10.3390/ph5010016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 01/23/2023] Open
Abstract
One approach to develop successful pain therapies is the modulation of dysfunctional ion channels that contribute to the detection of thermal, mechanical and chemical painful stimuli. These ion channels, known as thermoTRPs, promote the sensitization and activation of primary sensory neurons known as nociceptors. Pharmacological blockade and genetic deletion of thermoTRP have validated these channels as therapeutic targets for pain intervention. Several thermoTRP modulators have progressed towards clinical development, although most failed because of the appearance of unpredicted side effects. Thus, there is yet a need to develop novel channel modulators with improved therapeutic index. Here, we review the current state-of-the art and illustrate new pharmacological paradigms based on TRPV1 that include: (i) the identification of activity-dependent modulators of this thermoTRP channel; (ii) the design of allosteric modulators that interfere with protein-protein interaction involved in the functional coupling of stimulus sensing and gate opening; and (iii) the development of compounds that abrogate the inflammation-mediated increase of receptor expression in the neuronal surface. These new sites of action represent novel strategies to modulate pathologically active TRPV1, while minimizing an effect on the TRPV1 subpopulation involved in physiological and protective roles, thus increasing their potential therapeutic use.
Collapse
|
31
|
Fernández-Ballester G, Fernández-Carvajal A, González-Ros JM, Ferrer-Montiel A. Ionic channels as targets for drug design: a review on computational methods. Pharmaceutics 2011; 3:932-53. [PMID: 24309315 PMCID: PMC3857065 DOI: 10.3390/pharmaceutics3040932] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 10/26/2011] [Accepted: 11/30/2011] [Indexed: 01/21/2023] Open
Abstract
Ion channels are involved in a broad range of physiological and pathological processes. The implications of ion channels in a variety of diseases, including diabetes, epilepsy, hypertension, cancer and even chronic pain, have signaled them as pivotal drug targets. Thus far, drugs targeting ion channels were developed without detailed knowledge of the molecular interactions between the lead compounds and the target channels. In recent years, however, the emergence of high-resolution structures for a plethora of ion channels paves the way for computer-assisted drug design. Currently, available functional and structural data provide an attractive platform to generate models that combine substrate-based and protein-based approaches. In silico approaches include homology modeling, quantitative structure-activity relationships, virtual ligand screening, similarity and pharmacophore searching, data mining, and data analysis tools. These strategies have been frequently used in the discovery and optimization of novel molecules with enhanced affinity and specificity for the selected therapeutic targets. In this review we summarize recent applications of in silico methods that are being used for the development of ion channel drugs.
Collapse
|