1
|
Qi Y, Gong H, Shen Z, Wu L, Xu Z, Shi N, Lin K, Tian M, Xu Z, Li X, Zhao Q. TRPM8 and TRPA1 ideal targets for treating cold-induced pain. Eur J Med Chem 2025; 282:117043. [PMID: 39571458 DOI: 10.1016/j.ejmech.2024.117043] [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: 09/21/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 12/10/2024]
Abstract
TRP channels are essential for detecting variations in external temperature and are ubiquitously expressed in both the peripheral and central nervous systems as integral channel proteins. They primarily mediate a range of sensory responses, including thermal sensations, nociception, mechanosensation, vision, and gustation, thus playing a critical role in regulating various physiological functions. In colder climates, individuals often experience pain associated with low temperatures, leading to significant discomfort. Within the TRP channel family, TRPM8 and TRPA1 ion channels serve as the primary sensors for cold temperature fluctuations and are integral to both cold nociception and neuropathic pain pathways. Recent advancements in the biosynthesis of inhibitors targeting TRPM8 and TRPA1 have prompted the need for a comprehensive review of their structural characteristics, biological activities, biosynthetic pathways, and chemical synthesis. This paper aims to delineate the distinct roles of TRPM8 and TRPA1 in pain perception, elucidate their respective protein structures, and compile various combinations of TRPM8 and TRPA1 antagonists and agonists. The discussion encompasses their chemical structures, structure-activity relationships (SARs), biological activities, selectivity, and therapeutic potential, with a particular focus on the conformational relationships between antagonists and the channels. This review seeks to provide in-depth insights into pharmacological strategies for managing pain associated with TRPM8 and TRPA1 activation and will pave the way for future investigations into pharmacotherapeutic approaches for alleviating cold-induced pain.
Collapse
Affiliation(s)
- Yiming Qi
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Hao Gong
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Zixian Shen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Limeng Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Zonghe Xu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Nuo Shi
- College of Pharmacy, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Kexin Lin
- College of Pharmacy, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Meng Tian
- College of Pharmacy, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zihua Xu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China.
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China.
| |
Collapse
|
2
|
Behrendt M. Implications of TRPM3 and TRPM8 for sensory neuron sensitisation. Biol Chem 2024; 405:583-599. [PMID: 39417661 DOI: 10.1515/hsz-2024-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sensory neurons serve to receive and transmit a wide range of information about the conditions of the world around us as well as the external and internal state of our body. Sensitisation of these nerve cells, i.e. becoming more sensitive to stimuli or the emergence or intensification of spontaneous activity, for example in the context of inflammation or nerve injury, can lead to chronic diseases such as neuropathic pain. For many of these disorders there are only very limited treatment options and in order to find and establish new therapeutic approaches, research into the exact causes of sensitisation with the elucidation of the underlying mechanisms and the identification of the molecular components is therefore essential. These components include plasma membrane receptors and ion channels that are involved in signal reception and transmission. Members of the transient receptor potential (TRP) channel family are also expressed in sensory neurons and some of them play a crucial role in temperature perception. This review article focuses on the heat-sensitive TRPM3 and the cold-sensitive TRPM8 (and TRPA1) channels and their importance in sensitisation of dorsal root ganglion sensory neurons is discussed based on studies related to inflammation and injury- as well as chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Marc Behrendt
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, MCTN, Tridomus, Building C, Ludolf-Krehl-Straße 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
3
|
Gold MS, Pineda-Farias JB, Close D, Patel S, Johnston PA, Stocker SD, Journigan VB. Subcutaneous administration of a novel TRPM8 antagonist reverses cold hypersensitivity while attenuating the drop in core body temperature. Br J Pharmacol 2024; 181:3527-3543. [PMID: 38794851 DOI: 10.1111/bph.16429] [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: 07/17/2023] [Revised: 04/01/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND AND PURPOSE We extend the characterization of the TRPM8 antagonist VBJ103 with tests of selectivity, specificity and distribution, therapeutic efficacy of systemic administration against oxaliplatin-induced cold hyperalgesia and the impact of systemic administration on core body temperature (CBT). EXPERIMENTAL APPROACH Selectivity at human TRPA1 and TRPV1 as well as in vitro safety profiling was determined. Effects of systemic administration of VBJ103 were evaluated in a model of oxaliplatin-induced cold hyperalgesia. Both peripheral and centrally mediated effects of VBJ103 on CBT were assessed with radiotelemetry. KEY RESULTS VBJ103 had no antagonist activity at TRPV1 and TRPA1, but low potency TRPA1 activation. The only safety liability detected was partial inhibition of the dopamine transporter (DAT). VBJ103 delivered subcutaneously dose-dependently attenuated cold hypersensitivity in oxaliplatin-treated mice at 3, 10 and 30 mg·kg-1 (n = 7, P < 0.05). VBJ103 (30 mg·kg-1) antinociception was influenced by neither the TRPA1 antagonist HC-030031 nor the DAT antagonist GBR12909. Subcutaneous administration of VBJ103 (3, 10 and 30 mg·kg-1, but not 100 or 300 mg·kg-1, n = 7) decreased CBT (2°C). Intraperitoneal (i.p.) administration of VBJ103 (3, 10 and 30 mg·kg-1) dose-dependently decreased CBT to an extent larger than that detected with subcutaneous administration. Intracerebroventricular (i.c.v.) administration (306 nmol/1 μL; n = 5) did not alter CBT. CONCLUSIONS AND IMPLICATIONS We achieve therapeutic efficacy with subcutaneous administration of a novel TRPM8 antagonist that attenuates deleterious influences on CBT, a side effect that has largely prevented the translation of TRPM8 as a target.
Collapse
Affiliation(s)
- Michael S Gold
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jorge B Pineda-Farias
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David Close
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Smith Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sean D Stocker
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - V Blair Journigan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Petushkov VN, Vavilov MV, Khokhlova AN, Zagitova RI, Belozerova OA, Shcheglov AS, Kovalchuk SI, Tsarkova AS, Rodionova NS, Yampolsky IV, Dubinnyi MA. Henlea earthworm bioluminescence comprises violet-blue BRET from tryptophan 2-carboxylate to deazaflavin cofactor. Biochem Biophys Res Commun 2024; 708:149787. [PMID: 38537527 DOI: 10.1016/j.bbrc.2024.149787] [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: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
We recently identified the deazaflavin cofactor as a light emitter in novel bioluminescence (BL) system from Siberian earthworms Henlea sp. (Petushkov et al., 2023, Org. Biomol. Chem. 21:415-427). In the present communication we compared in vitro BL spectra in the absence and in the presence of the cofactor and found a wavelength shift from 420 to 476 nm. This violet-blue BRET to deazaflavin cofactor (acceptor of photonless transfer) masks the actual oxyluciferin as an emitter (BRET donor) in the novel BL system. The best candidate for that masked chromophore is tryptophan 2-carboxylate (T2C) found previously as a building block in some natural products isolated from Henlea sp. (Dubinnyi et al., 2020, ChemSelect 5:13155-13159). We synthesized T2C and acetyl-T2C, verified their presence in earthworms by nanoflow-HRMS, explored spectral properties of excitation and emission spectra and found a chain of excitation/emission maxima with a perfect potential for BRET: 300 nm (excitation of T2C) - 420 nm (emission of T2C) - 420 nm (excitation of deazaflavin) - 476 nm (emission of deazaflavin, BL). An array of natural products with T2C chromophore are present in BL earthworms as candidates for novel oxyluciferin. We demonstrated for the Henlea BL that the energy of the excited state of the T2C chromophore is transferred by the Förster mechanism and then emitted by deazaflavin (BRET), similarly to known examples: aequorin-GFP in Aequorea victoria and antenna proteins in bacterial BL systems (lumazine from Photobacterium and yellow fluorescent protein from Vibrio fischeri strain Y1).
Collapse
Affiliation(s)
- Valentin N Petushkov
- Institute of Biophysics, Krasnoyarsk Research Center, Siberian Branch, Russian Academy of Sciences, 50/50 Akademgorodok, 660036, Krasnoyarsk, Russia.
| | - Matvey V Vavilov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia; Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33k4 Profsoyuznaya str., Moscow, 117418, Russia
| | - Anastasia N Khokhlova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia; Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1 bld. 3, Moscow, 119991у, Russia
| | - Renata I Zagitova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia
| | - Olga A Belozerova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia
| | - Aleksandr S Shcheglov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia; Pirogov Russian National Research Medical University, 1 Ostrovityanova str, Moscow, 117997, Russia
| | - Sergey I Kovalchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia
| | - Aleksandra S Tsarkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia
| | - Natalia S Rodionova
- Institute of Biophysics, Krasnoyarsk Research Center, Siberian Branch, Russian Academy of Sciences, 50/50 Akademgorodok, 660036, Krasnoyarsk, Russia
| | - Ilia V Yampolsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia; Pirogov Russian National Research Medical University, 1 Ostrovityanova str, Moscow, 117997, Russia
| | - Maxim A Dubinnyi
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya str., Moscow, 117997, Russia; Moscow Institute of Physics and Technology (State University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141700, Russia.
| |
Collapse
|
5
|
Martín-Escura C, Bonache MÁ, Medina JA, Medina-Peris A, De Andrés-López J, González-Rodríguez S, Kerselaers S, Fernández-Ballester G, Voets T, Ferrer-Montiel A, Fernández-Carvajal A, González-Muñiz R. β-Lactam TRPM8 Antagonists Derived from Phe-Phenylalaninol Conjugates: Structure-Activity Relationships and Antiallodynic Activity. Int J Mol Sci 2023; 24:14894. [PMID: 37834342 PMCID: PMC10573892 DOI: 10.3390/ijms241914894] [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: 08/31/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
The protein transient receptor potential melastatin type 8 (TRPM8), a non-selective, calcium (Ca2+)-permeable ion channel is implicated in several pathological conditions, including neuropathic pain states. In our previous research endeavors, we have identified β-lactam derivatives with high hydrophobic character that exhibit potent and selective TRPM8 antagonist activity. This work describes the synthesis of novel derivatives featuring C-terminal amides and diversely substituted N'-terminal monobenzyl groups in an attempt to increase the total polar surface area (TPSA) in this family of compounds. The primary goal was to assess the influence of these substituents on the inhibition of menthol-induced cellular Ca2+ entry, thereby establishing critical structure-activity relationships. While the substitution of the tert-butyl ester by isobutyl amide moieties improved the antagonist activity, none of the N'-monobencyl derivatives, regardless of the substituent on the phenyl ring, achieved the activity of the model dibenzyl compound. The antagonist potency of the most effective compounds was subsequently verified using Patch-Clamp electrophysiology experiments. Furthermore, we evaluated the selectivity of one of these compounds against other members of the transient receptor potential (TRP) ion channel family and some receptors connected to peripheral pain pathways. This compound demonstrated specificity for TRPM8 channels. To better comprehend the potential mode of interaction, we conducted docking experiments to uncover plausible binding sites on the functionally active tetrameric protein. While the four main populated poses are located by the pore zone, a similar location to that described for the N-(3-aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)-benzamide (AMTB) antagonist cannot be discarded. Finally, in vivo experiments, involving a couple of selected compounds, revealed significant antinociceptive activity within a mice model of cold allodynia induced by oxaliplatin (OXA).
Collapse
Affiliation(s)
- Cristina Martín-Escura
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain
- Alodia Farmacéutica SL, 28108 Alcobendas, Spain
| | | | | | | | | | - Sara González-Rodríguez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain
- Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Julián Clavería 6, 33006 Oviedo, Spain
| | - Sara Kerselaers
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain and Disease Research, KU Leuven, Herestraat 49 Box 802, 3000 Leuven, Belgium
| | | | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain and Disease Research, KU Leuven, Herestraat 49 Box 802, 3000 Leuven, Belgium
| | | | | | | |
Collapse
|
6
|
In Silico Identification and In Vitro Evaluation of New ABCG2 Transporter Inhibitors as Potential Anticancer Agents. Int J Mol Sci 2022; 24:ijms24010725. [PMID: 36614168 PMCID: PMC9820944 DOI: 10.3390/ijms24010725] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Different molecular mechanisms contribute to the development of multidrug resistance in cancer, including increased drug efflux, enhanced cellular repair mechanisms and alterations of drug metabolism or drug targets. ABCG2 is a member of the ATP-binding cassette superfamily transporters that promotes drug efflux, inducing chemotherapeutic resistance in malignant cells. In this context, the development of selective ABCG2 inhibitors might be a suitable strategy to improve chemotherapy efficacy. Thus, through a multidisciplinary approach, we identified a new ABCG2 selective inhibitor (8), highlighting its ability to increase mitoxantrone cytotoxicity in both hepatocellular carcinoma (EC50from 8.67 ± 2.65 to 1.25 ± 0.80 μM) and transfected breast cancer cell lines (EC50from 9.92 ± 2.32 to 2.45 ± 1.40 μM). Moreover, mitoxantrone co-administration in both transfected and non-transfected HEK293 revealed that compound 8 notably lowered the mitoxantrone EC50, demonstrating its efficacy along with the importance of the ABCG2 extrusion pump overexpression in MDR reversion. These results were corroborated by evaluating the effect of inhibitor 8 on mitoxantrone cell uptake in multicellular tumor spheroids and via proteomic experiments.
Collapse
|
7
|
Di Sarno V, Giovannelli P, Medina-Peris A, Ciaglia T, Di Donato M, Musella S, Lauro G, Vestuto V, Smaldone G, Di Matteo F, Bifulco G, Castoria G, Migliaccio A, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, Ostacolo C, Bertamino A. New TRPM8 blockers exert anticancer activity over castration-resistant prostate cancer models. Eur J Med Chem 2022; 238:114435. [DOI: 10.1016/j.ejmech.2022.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/04/2022]
|
8
|
Martín-Escura C, Medina-Peris A, Spear LA, de la Torre Martínez R, Olivos-Oré LA, Barahona MV, González-Rodríguez S, Fernández-Ballester G, Fernández-Carvajal A, Artalejo AR, Ferrer-Montiel A, González-Muñiz R. β-Lactam TRPM8 Antagonist RGM8-51 Displays Antinociceptive Activity in Different Animal Models. Int J Mol Sci 2022; 23:ijms23052692. [PMID: 35269831 PMCID: PMC8910920 DOI: 10.3390/ijms23052692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential melastatin subtype 8 (TRPM8) is a cation channel extensively expressed in sensory neurons and implicated in different painful states. However, the effectiveness of TRPM8 modulators for pain relief is still a matter of discussion, since structurally diverse modulators lead to different results, depending on the animal pain model. In this work, we described the antinociceptive activity of a β–lactam derivative, RGM8-51, showing good TRPM8 antagonist activity, and selectivity against related thermoTRP channels and other pain-mediating receptors. In primary cultures of rat dorsal root ganglion (DRG) neurons, RGM8-51 potently reduced menthol-evoked neuronal firing without affecting the major ion conductances responsible for action potential generation. This compound has in vivo antinociceptive activity in response to cold, in a mouse model of oxaliplatin-induced peripheral neuropathy. In addition, it reduces cold, mechanical and heat hypersensitivity in a rat model of neuropathic pain arising after chronic constriction of the sciatic nerve. Furthermore, RGM8-51 exhibits mechanical hypersensitivity-relieving activity, in a mouse model of NTG-induced hyperesthesia. Taken together, these preclinical results substantiate that this TRPM8 antagonist is a promising pharmacological tool to study TRPM8-related diseases.
Collapse
Affiliation(s)
- Cristina Martín-Escura
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Alodia Farmacéutica SL, 28108 Alcobendas, Spain
| | - Alicia Medina-Peris
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luke A. Spear
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
| | - Roberto de la Torre Martínez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luis A. Olivos-Oré
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Victoria Barahona
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sara González-Rodríguez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Gregorio Fernández-Ballester
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Asia Fernández-Carvajal
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| | - Antonio R. Artalejo
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonio Ferrer-Montiel
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Rosario González-Muñiz
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| |
Collapse
|
9
|
Iraci N, Ostacolo C, Medina-Peris A, Ciaglia T, Novoselov AM, Altieri A, Cabañero D, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, Bertamino A, Kurkin AV. In Vitro and In Vivo Pharmacological Characterization of a Novel TRPM8 Inhibitor Chemotype Identified by Small-Scale Preclinical Screening. Int J Mol Sci 2022; 23:2070. [PMID: 35216186 PMCID: PMC8877448 DOI: 10.3390/ijms23042070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
Transient receptor potential melastatin type 8 (TRPM8) is a target for the treatment of different physio-pathological processes. While TRPM8 antagonists are reported as potential drugs for pain, cancer, and inflammation, to date only a limited number of chemotypes have been investigated and thus a limited number of compounds have reached clinical trials. Hence there is high value in searching for new TRPM8 antagonistic to broaden clues to structure-activity relationships, improve pharmacological properties and explore underlying molecular mechanisms. To address this, the EDASA Scientific in-house molecular library has been screened in silico, leading to identifying twenty-one potentially antagonist compounds of TRPM8. Calcium fluorometric assays were used to validate the in-silico hypothesis and assess compound selectivity. Four compounds were identified as selective TRPM8 antagonists, of which two were dual-acting TRPM8/TRPV1 modulators. The most potent TRPM8 antagonists (BB 0322703 and BB 0322720) underwent molecular modelling studies to highlight key structural features responsible for drug-protein interaction. The two compounds were also investigated by patch-clamp assays, confirming low micromolar potencies. The most potent compound (BB 0322703, IC50 1.25 ± 0.26 μM) was then profiled in vivo in a cold allodinya model, showing pharmacological efficacy at 30 μM dose. The new chemotypes identified showed remarkable pharmacological properties paving the way to further investigations for drug discovery and pharmacological purposes.
Collapse
Affiliation(s)
- Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy;
| | - Carmine Ostacolo
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (C.O.); (I.G.-M.)
| | - Alicia Medina-Peris
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Anton M. Novoselov
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
| | - Andrea Altieri
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
- EDASA Scientific srls, Via Stingi 37, 66050 San Salvo, Italy
| | - David Cabañero
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Asia Fernandez-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Isabel Gomez-Monterrey
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (C.O.); (I.G.-M.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Alexander V. Kurkin
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
| |
Collapse
|
10
|
A review of synthetic bioactive tetrahydro-β-carbolines: A medicinal chemistry perspective. Eur J Med Chem 2021; 225:113815. [PMID: 34479038 DOI: 10.1016/j.ejmech.2021.113815] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/22/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022]
Abstract
1, 2, 3, 4-Tetrahydro-β-carboline (THβC) scaffold is widespread in many natural products (NPs) and synthetic compounds which show a variety of pharmacological activities. In this article, we reviewed the design, structures and biological characteristics of reported synthetic THβC compounds, and structure and activity relationship (SAR) of them were also discussed. This work might provide a reference for subsequent drug development based on THβC.
Collapse
|
11
|
Di Donato M, Ostacolo C, Giovannelli P, Di Sarno V, Monterrey IMG, Campiglia P, Migliaccio A, Bertamino A, Castoria G. Therapeutic potential of TRPM8 antagonists in prostate cancer. Sci Rep 2021; 11:23232. [PMID: 34853378 PMCID: PMC8636514 DOI: 10.1038/s41598-021-02675-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential melastatin-8 (TRPM8) represents an emerging target in prostate cancer, although its mechanism of action remains unclear. Here, we have characterized and investigated the effects of TRPM8 modulators in prostate cancer aggressiveness disclosing the molecular mechanism underlying their biological activity. Patch-clamp and calcium fluorometric assays were used to characterize the synthesized compounds. Androgen-stimulated prostate cancer-derived cells were challenged with the compounds and the DNA synthesis was investigated in a preliminary screening. The most effective compounds were then employed to inhibit the pro-metastatic behavior of in various PC-derived cells, at different degree of malignancy. The effect of the compounds was then assayed in prostate cancer cell-derived 3D model and the molecular targets of selected compounds were lastly identified using transcriptional and non-transcriptional reporter assays. TRPM8 antagonists inhibit the androgen-dependent prostate cancer cell proliferation, migration and invasiveness. They are highly effective in reverting the androgen-induced increase in prostate cancer cell spheroid size. The compounds also revert the proliferation of castrate-resistant prostate cancer cells, provided they express the androgen receptor. In contrast, no effects were recorded in prostate cancer cells devoid of the receptor. Selected antagonists interfere in non-genomic androgen action and abolish the androgen-induced androgen receptor/TRPM8 complex assembly as well as the increase in intracellular calcium levels in prostate cancer cells. Our results shed light in the processes controlling prostate cancer progression and make the transient receptor potential melastatin-8 as a ‘druggable’ target in the androgen receptor-expressing prostate cancers.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131, Naples, Italy
| | - Pia Giovannelli
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy
| | - Isabel M Gomez Monterrey
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131, Naples, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy.
| | - Gabriella Castoria
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
12
|
Gulezian E, Crivello C, Bednenko J, Zafra C, Zhang Y, Colussi P, Hussain S. Membrane protein production and formulation for drug discovery. Trends Pharmacol Sci 2021; 42:657-674. [PMID: 34270922 DOI: 10.1016/j.tips.2021.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Integral membrane proteins (MPs) are important drug targets across most fields of medicine, but historically have posed a major challenge for drug discovery due to difficulties in producing them in functional forms. We review the state of the art in drug discovery strategies using recombinant multipass MPs, and outline methods to successfully express, stabilize, and formulate them for small-molecule and monoclonal antibody therapeutics development. Advances in structure-based drug design and high-throughput screening are allowing access to previously intractable targets such as ion channels and transporters, propelling the field towards the development of highly specific therapies targeting desired conformations.
Collapse
Affiliation(s)
- Ellen Gulezian
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | | | - Janna Bednenko
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Claudia Zafra
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Yihui Zhang
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Paul Colussi
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Sunyia Hussain
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA.
| |
Collapse
|
13
|
Izquierdo C, Martín-Martínez M, Gómez-Monterrey I, González-Muñiz R. TRPM8 Channels: Advances in Structural Studies and Pharmacological Modulation. Int J Mol Sci 2021; 22:ijms22168502. [PMID: 34445208 PMCID: PMC8395166 DOI: 10.3390/ijms22168502] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential melastatin subtype 8 (TRPM8) is a cold sensor in humans, activated by low temperatures (>10, <28 °C), but also a polymodal ion channel, stimulated by voltage, pressure, cooling compounds (menthol, icilin), and hyperosmolarity. An increased number of experimental results indicate the implication of TRPM8 channels in cold thermal transduction and pain detection, transmission, and maintenance in different tissues and organs. These channels also have a repercussion on different kinds of life-threatening tumors and other pathologies, which include urinary and respiratory tract dysfunctions, dry eye disease, and obesity. This compendium firstly covers newly described papers on the expression of TRPM8 channels and their correlation with pathological states. An overview on the structural knowledge, after cryo-electron microscopy success in solving different TRPM8 structures, as well as some insights obtained from mutagenesis studies, will follow. Most recently described families of TRPM8 modulators are also covered, along with a section of molecules that have reached clinical trials. To finalize, authors provide an outline of the potential prospects in the TRPM8 field.
Collapse
Affiliation(s)
- Carolina Izquierdo
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Programa de Doctorado en Química Orgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mercedes Martín-Martínez
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (I.G.-M.); (R.G.-M.)
| | - Rosario González-Muñiz
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Correspondence: (I.G.-M.); (R.G.-M.)
| |
Collapse
|
14
|
Phenylalanine-Derived β-Lactam TRPM8 Modulators. Configuration Effect on the Antagonist Activity. Int J Mol Sci 2021; 22:ijms22052370. [PMID: 33673444 PMCID: PMC7956626 DOI: 10.3390/ijms22052370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential cation channel subfamily M member 8 (TRPM8) is a Ca2+ non-selective ion channel implicated in a variety of pathological conditions, including cancer, inflammatory and neuropathic pain. In previous works we identified a family of chiral, highly hydrophobic β–lactam derivatives, and began to intuit a possible effect of the stereogenic centers on the antagonist activity. To investigate the influence of configuration on the TRPM8 antagonist properties, here we prepare and characterize four possible diastereoisomeric derivatives of 4-benzyl-1-[(3′-phenyl-2′-dibenzylamino)prop-1′-yl]-4-benzyloxycarbonyl-3-methyl-2-oxoazetidine. In microfluorography assays, all isomers were able to reduce the menthol-induced cell Ca2+ entry to larger or lesser extent. Potency follows the order 3R,4R,2′R > 3S,4S,2′R ≅ 3R,4R,2′S > 3S,4S,2′S, with the most potent diastereoisomer showing a half inhibitory concentration (IC50) in the low nanomolar range, confirmed by Patch-Clamp electrophysiology experiments. All four compounds display high receptor selectivity against other members of the TRP family. Furthermore, in primary cultures of rat dorsal root ganglion (DRG) neurons, the most potent diastereoisomers do not produce any alteration in neuronal excitability, indicating their high specificity for TRPM8 channels. Docking studies positioned these β-lactams at different subsites by the pore zone, suggesting a different mechanism than the known N-(3-aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)-benzamide (AMTB) antagonist.
Collapse
|
15
|
Kobayashi JI, Hirasawa H, Fujimori Y, Nakanishi O, Kamada N, Ikeda T, Yamamoto A, Kanbe H. Identification of N-acyl-N-indanyl-α-phenylglycinamides as selective TRPM8 antagonists designed to mitigate the risk of adverse effects. Bioorg Med Chem 2020; 30:115903. [PMID: 33333445 DOI: 10.1016/j.bmc.2020.115903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/26/2020] [Indexed: 01/09/2023]
Abstract
Transient receptor potential melastatin 8 (TRPM8), a temperature-sensitive ion channel responsible for detecting cold, is an attractive molecular target for the treatment of pain and other disorders. We have previously discovered a selective TRPM8 antagonist, KPR-2579, which inhibited bladder afferent hyperactivity induced by acetic acid instillation into the bladder. However, additional studies have revealed potential adverse effects with KPR-2579, such as the formation of a reactive metabolite, CYP3A4 induction, and convulsions. In this report, we describe the optimization of α-phenylglycinamide derivatives to mitigate the risk of these adverse effects. The optimal compound 13x exhibited potent inhibition against icilin-induced wet-dog shakes and cold-induced frequent voiding in rats, with a wide safety margin against the potential side effects.
Collapse
Affiliation(s)
- Jun-Ichi Kobayashi
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan.
| | - Hideaki Hirasawa
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Yoshikazu Fujimori
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Osamu Nakanishi
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Noboru Kamada
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Tetsuya Ikeda
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Akitoshi Yamamoto
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| | - Hiroki Kanbe
- Discovery Research, R&D, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka kashiwabara, Azumino, Nagano 399-8304, Japan
| |
Collapse
|
16
|
Leng HJ, Wang YT, He XH, Xia HL, Xu PS, Xiang P, He QQ, Zhan G, Huang W. Design and Efficient Synthesis of RalA Inhibitors Containing the Dihydro-α-carboline Scaffold. ChemMedChem 2020; 16:851-859. [PMID: 33244883 DOI: 10.1002/cmdc.202000722] [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: 09/12/2020] [Revised: 11/02/2020] [Indexed: 11/07/2022]
Abstract
Ras-related protein RalA is a member of the Ras small GTPases superfamily. Its activation plays an important role in regulating tumor initiation, invasion, migration, and metastasis. In this study, we designed a new type of RalA inhibitor containing a dihydro-α-carboline scaffold. The structurally new dihydro-α-carboline derivatives could be efficiently synthesized in good yields through a newly developed three-component [3+2+1] cyclization reaction. Evaluation of the biological activity showed that some of the dihydro-α-carboline derivatives can inhibit RalA/B and proliferative activities of NSCLC cell lines. The 4-(pyridin-3-yl)-dihydro-α-carboline compound (3 o) was found to be the most potent derivative, with IC50 values of 0.43±0.03, 0.64±0.07, 0.93±0.10, and 1.54±0.15 μM against A549, H1299, H460, and H1975 cells, respectively. Mechanism investigation suggested that 3 o inhibits the RalA/B activation of A549, down-regulates Bcl-2, stimulates cytochrome c and PARP cleavage, and induces cell apoptosis. A molecular docking study revealed that 3 o can form stable hydrogen bonds with residues of RalA. Moreover, amide-π and alkyl-π interactions also contributed to the affinity between 3 o and RalA.
Collapse
Affiliation(s)
- Hai-Jun Leng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.,Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, 610052, Chengdu, China
| | - Yu-Ting Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Xiang-Hong He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Hou-Lin Xia
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Peng-Shuai Xu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, 610052, Chengdu, China
| | - Peng Xiang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, 610052, Chengdu, China
| | - Qing-Qing He
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, 610052, Chengdu, China
| | - Gu Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| |
Collapse
|