Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 394
Filtrar
1.
Cell Rep ; 43(4): 114108, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38615321

RESUMEN

TRP channels are implicated in various diseases, but high structural similarity between them makes selective pharmacological modulation challenging. Here, we study the molecular mechanism underlying specific inhibition of the TRPM7 channel, which is essential for cancer cell proliferation, by the anticancer agent CCT128930 (CCT). Using cryo-EM, functional analysis, and MD simulations, we show that CCT binds to a vanilloid-like (VL) site, stabilizing TRPM7 in the closed non-conducting state. Similar to other allosteric inhibitors of TRPM7, NS8593 and VER155008, binding of CCT is accompanied by displacement of a lipid that resides in the VL site in the apo condition. Moreover, we demonstrate the principal role of several residues in the VL site enabling CCT to inhibit TRPM7 without impacting the homologous TRPM6 channel. Hence, our results uncover the central role of the VL site for the selective interaction of TRPM7 with small molecules that can be explored in future drug design.


Asunto(s)
1-Naftilamina/análogos & derivados , Antineoplásicos , Canales Catiónicos TRPM , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células HEK293 , Simulación de Dinámica Molecular , Sitios de Unión , Unión Proteica , Microscopía por Crioelectrón
2.
J Nat Prod ; 87(4): 783-797, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38537009

RESUMEN

Waixenicin A, a xenicane diterpene from the octocoral Sarcothelia edmondsoni, is a selective, potent inhibitor of the TRPM7 ion channel. To study the structure-activity relationship (SAR) of waixenicin A, we isolated and assayed related diterpenes from S. edmondsoni. In addition to known waixenicins A (1) and B (2), we purified six xenicane diterpenes, 7S,8S-epoxywaixenicins A (3) and B (4), 12-deacetylwaixenicin A (5), waixenicin E (6), waixenicin F (7), and 20-acetoxyxeniafaraunol B (8). We elucidated the structures of 3-8 by NMR and MS analyses. Compounds 1, 2, 3, 4, and 6 inhibited TRPM7 activity in a cell-based assay, while 5, 7, and 8 were inactive. A preliminary SAR emerged showing that alterations to the nine-membered ring of 1 did not reduce activity, while the 12-acetoxy group, in combination with the dihydropyran, appears to be necessary for TRPM7 inhibition. The bioactive compounds are proposed to be latent electrophiles by formation of a conjugated oxocarbenium ion intermediate. Whole-cell patch-clamp experiments demonstrated that waixenicin A inhibition is irreversible, consistent with a covalent inhibitor, and showed nanomolar potency for waixenicin B (2). Conformational analysis (DFT) of 1, 3, 7, and 8 revealed insights into the conformation of waixenicin A and congeners and provided information regarding the stabilization of the proposed pharmacophore.


Asunto(s)
Acetatos , Antozoos , Diterpenos , Proteínas Serina-Treonina Quinasas , Canales Catiónicos TRPM , Diterpenos/farmacología , Diterpenos/química , Diterpenos/aislamiento & purificación , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Antozoos/química , Relación Estructura-Actividad , Humanos , Estructura Molecular , Hawaii , Conformación Molecular
3.
J Nat Prod ; 87(4): 722-732, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38408345

RESUMEN

The first detailed phytochemical analysis of the cannabigerol (CBG)-rich chemotype IV of Cannabis sativa L. resulted in the isolation of the expected cannabigerolic acid/cannabigerol (CBGA/CBG) and cannabidiolic acid/cannabidiol (CBDA/CBD) and of nine new phytocannabinoids (5-13), which were fully characterized by HR-ESIMS and 1D and 2D NMR. These included mono- or dihydroxylated CBGA/CBG analogues, a congener with a truncated side chain (10), cyclocannabigerol B (11), and the CBD derivatives named cannabifuranols (12 and 13). Cyclocannabigerol B and cannabifuranols are characterized by a novel phytocannabinoid structural architecture. The isolated phytocannabinoids were assayed on the receptor channels TRPA1 and TRPM8, unveiling a potent dual TRPA1 agonist/TRPM8 antagonist profile for compounds 6, 7, and 14. Chiral separation of the two enantiomers of 5 resulted in the discovery of a synergistic effect of the two enantiomers on TRPA1.


Asunto(s)
Cannabinoides , Cannabis , Canal Catiónico TRPA1 , Canales Catiónicos TRPM , Canales de Potencial de Receptor Transitorio , Cannabis/química , Canal Catiónico TRPA1/antagonistas & inhibidores , Cannabinoides/farmacología , Cannabinoides/química , Cannabinoides/aislamiento & purificación , Canales Catiónicos TRPM/antagonistas & inhibidores , Estructura Molecular , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Fitoquímicos/farmacología , Fitoquímicos/aislamiento & purificación , Fitoquímicos/química , Humanos , Cannabidiol/farmacología , Cannabidiol/química , Canales de Calcio/metabolismo
4.
Function (Oxf) ; 5(1): zqad069, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38162115

RESUMEN

Cannabinoids are a major class of compounds produced by the plant Cannabis sativa. Previous work has demonstrated that the main cannabinoids cannabidiol (CBD) and tetrahydrocannabinol (THC) can have some beneficial effects on pain, inflammation, epilepsy, and chemotherapy-induced nausea and vomiting. While CBD and THC represent the two major plant cannabinoids, some hemp varieties with enzymatic deficiencies produce mainly cannabigerolic acid (CBGA). We recently reported that CBGA has a potent inhibitory effect on both Store-Operated Calcium Entry (SOCE) via inhibition of Calcium Release-Activated Calcium (CRAC) channels as well as currents carried by the channel-kinase TRPM7. Importantly, CBGA prevented kidney damage and suppressed mRNA expression of inflammatory cytokines through inhibition of these mechanisms in an acute nephropathic mouse model. In the present study, we investigate the most common major and minor cannabinoids to determine their potential efficacy on TRPM7 channel function. We find that approximately half of the tested cannabinoids suppress TRPM7 currents to some degree, with CBGA having the strongest inhibitory effect on TRPM7. We determined that the CBGA-mediated inhibition of TRPM7 requires a functional kinase domain, is sensitized by both intracellular Mg⋅ATP and free Mg2+ and reduced by increases in intracellular Ca2+. Finally, we demonstrate that CBGA inhibits native TRPM7 channels in a B lymphocyte cell line. In conclusion, we demonstrate that CBGA is the most potent cannabinoid in suppressing TRPM7 activity and possesses therapeutic potential for diseases in which TRPM7 is known to play an important role such as cancer, stroke, and kidney disease.


Asunto(s)
Cannabinoides , Canales Catiónicos TRPM , Animales , Ratones , Cannabinoides/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores
5.
Br J Pharmacol ; 180(11): 1482-1499, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36549668

RESUMEN

BACKGROUND AND PURPOSE: Interstitial cystitis (=painful bladder syndrome) is a chronic bladder syndrome characterised by pelvic and bladder pain, urinary frequency and urgency, and nocturia. Transient receptor potential (TRP) channels are an attractive target in reducing the pain associated with interstitial cystitis. The current study aims to determine the efficacy of combination of TRP vanilloid 1 (TRPV1) and TRP melastatin 8 (TRPM8) channel inhibition in reducing the pain associated with experimental cystitis in guinea pigs. EXPERIMENTAL APPROACH: A novel animal model of non-ulcerative interstitial cystitis has been developed using protamine sulfate/zymosan in female guinea pigs. Continuous voiding cystometry was performed in conscious guinea pigs. Ex vivo "close-to-target" single unit extracellular recordings were made from fine branches of pelvic nerves entering the guinea pig bladder. Visceromotor responses in vivo were used to determine the effects of TRP channel antagonists on cystitis-induced bladder hypersensitivity. KEY RESULTS: Protamine sulfate/zymosan treatment evoked mild inflammation in the bladder and increased micturition frequency in conscious animals. In cystitis, high threshold muscular afferents were sensitised via up-regulation of TRPV1 channels, high threshold muscular-mucosal afferents were sensitised via TRPM8 channels, and mucosal afferents by both. Visceromotor responses evoked by noxious bladder distension were significantly enhanced in cystitis and were returned to control levels upon administration of combination of low doses of TRPV1 and TRPM8 antagonists. CONCLUSIONS AND IMPLICATIONS: The data demonstrate the therapeutic promises of combination of TRPV1 and TRPM8 antagonists for the treatment of bladder hypersensitivity in cystitis.


Asunto(s)
Cistitis Intersticial , Cistitis , Hipersensibilidad , Canales de Potencial de Receptor Transitorio , Animales , Femenino , Cobayas , Cistitis/tratamiento farmacológico , Dolor , Canales Catiónicos TRPV , Vejiga Urinaria , Zimosan/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores
6.
Int J Oncol ; 60(4)2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35234266

RESUMEN

Melanoma continues to be the most aggressive and devastating form of skin cancer for which the development of novel therapies is required. The present study aimed to determine the effects of antagonism of the transient receptor potential melastatin­2 (TRPM2) ion channel in primary human malignant melanoma cells. TRPM2 antagonism via use of the antifungal agent, clotrimazole, led to decreases in cell proliferation, as well as dose­dependent increases in cell death in all melanoma cell lines investigated. The targeting of TRPM2 channels was verified using TRPM2 knockdown, where treatment with TRPM2 small­interfering RNA led to similar levels of cell death in all melanoma cell lines when compared with clotrimazole treatment. Minimal effects on proliferation and cell death were observed following antagonism or knockdown of TRPM2 in non­cancerous human keratinocytes. Moreover, characteristics of TRPM2 were explored in these melanoma cells and the results demonstrated that TRPM2, localized to the plasma membrane as a non­specific ion channel in non­cancerous cells, displayed a nuclear localization in all human melanoma cell lines analyzed. Additional characterization of these melanoma cell lines confirmed that each expressed one or more established multidrug resistance genes. Results of the present study therefore indicated that antagonism of the TRPM2 channel led to antitumor effects in human melanoma cells, including those that are potentially unresponsive to current treatments due to the expression of drug resistance genes. The unique cellular localization of TRPM2 and the specificity of the antitumor effects elicited by TRPM2 antagonism suggested that TRPM2 possesses a unique role in melanoma cells. Collectively, the targeting of TRPM2 represents a potentially novel, efficacious and readily accessible treatment option for patients with melanoma.


Asunto(s)
Línea Celular Tumoral/metabolismo , Melanoma/genética , Melanoma/prevención & control , Canales Catiónicos TRPM/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral/fisiología , Proliferación Celular/efectos de los fármacos , Humanos , Melanoma/tratamiento farmacológico
7.
Int J Mol Sci ; 23(4)2022 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-35216186

RESUMEN

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.


Asunto(s)
Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Descubrimiento de Drogas/métodos , Femenino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
8.
Metab Brain Dis ; 37(3): 711-728, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34989943

RESUMEN

The overload cytosolic free Ca2+ (cCa2+) influx-mediated excessive generation of oxidative stress in the pathophysiological conditions induces neuronal and cellular injury via the activation of cation channels. TRPM2 and TRPV4 channels are activated by oxidative stress, and their specific antagonists have not been discovered yet. The antioxidant and anti-Covid-19 properties of carvacrol (CARV) were recently reported. Hence, I suspected possible antagonist properties of CARV against oxidative stress (OS)/ADP-ribose (ADPR)-induced TRPM2 and GSK1016790A (GSK)-mediated TRPV4 activations in neuronal and kidney cells. I investigated the antagonist role of CARV on the activations of TRPM2 and TRPV4 in SH-SY5Y neuronal, BV-2 microglial, and HEK293 cells. The OS/ADPR and GSK in the cells caused to increase of TRPM2/TRPV4 current densities and overload cytosolic free Ca2+ (cCa2+) influx with an increase of mitochondrial membrane potential, cytosolic (cROS), and mitochondrial (mROS) ROS. The changes were not observed in the absence of TRPM2 and TRPV4 or the presence of Ca2+ free extracellular buffer and PARP-1 inhibitors (PJ34 and DPQ). When OS-induced TRPM2 and GSK-induced TRPV4 activations were inhibited by the treatment of CARV, the increase of cROS, mROS, lipid peroxidation, apoptosis, cell death, cCa2+ concentration, caspase -3, and caspase -9 levels were restored via upregulation of glutathione and glutathione peroxidase. In conclusion, the treatment of CARV modulated the TRPM2 and TRPV4-mediated overload Ca2+ influx and may provide an avenue for protecting TRPM2 and TRPV4-mediated neurodegenerative diseases associated with the increase of mROS and cCa2+. The possible TRPM2 and TRPV4 blocker action of carvacrol (CARV) via the modulation oxidative stress and apoptosis in the SH-SY5Y neuronal cells. TRPM2 is activated by DNA damage-induced (via PARP-1 activation) ADP-ribose (ADPR) and reactive oxygen species (ROS) (H2O2), although it is inhibited by nonspecific inhibitors (ACA and 2-APB). TRPV4 is activated by the treatments of GSK1016790A (GSK), although it is inhibited by a nonspecific inhibitor (ruthenium red, RuRe). The treatment of GSK induces excessive generation of ROS. The accumulation of free cytosolic Ca2+ (cCa2+) via the activations of TRPM2 and TRPV4 in the mitochondria causes the increase of mitochondrial membrane depolarization (ΔΨm). In turn, the increase of ΔΨm causes the excessive generation of ROS. The TRPM2 and TRPV4-induced the excessive generations of ROS result in the increase of apoptosis and cell death via the activations of caspase -3 (Casp-3) and caspase -9 (Casp-9) in the neuronal cells, although their oxidant actions decrease the glutathione (GSH) and glutathione peroxidase (GSHPx) levels. The oxidant and apoptotic adverse actions of TRPM2 and TRPV4 are modulated by the treatment of CARV.


Asunto(s)
Antioxidantes/farmacología , Cimenos/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPV/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno
9.
Behav Brain Res ; 423: 113765, 2022 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-35063497

RESUMEN

The roles of ion channels, miRNAs and, neurotransmitters in the pathophysiology of major depressive disorder (MDD) are not yet fully elucidated. The current study aims to investigate ion channel gene expressions in the brain, the therapeutic efficacies of TRPC1, TRPM4, and CHRNA6 inhibitors, miRNAs specific to these ion channels and, neurotransmitter interactions in a chronic unpredictable mild stress (CUMS) induced MDD rat model. 48 two-month-old male albino Wistar rats were divided into Control, CUMS, Sham, CUMS+Pico145 (TRPC1 inhibitor), CUMS+ 9-Phe (TRPM4 inhibitor), and CUMS+BPiDl (CHRNA6 inhibitor) groups. Seven-week CUMS was used to induce MDD. Inhibitors were administered subacutely on the final of CUMS. Rats were subjected to behavioral tests. Gene expression levels were analyzed using qRT-PCR and neurotransmitter levels using ELISA. CUMS lead to a significant upregulation in the expression of channels in the hippocampus, and channels in the prefrontal cortex. Behavioral experiments determined the antidepressant effects as follows: Pico145 > BPiDl > 9-Phe. Compared to the Control, serotonin and noradrenaline levels remained unchanged, whereas dopamine levels increased. Acetylcholine levels decreased in CUMS and CUMS+Pico145 groups. CUMS significantly altered the expression of 6 miRNAs in the brain. BPiDl upregulated the expression of miR-6334 and Pico145 upregulated the expression of miR-135b-5p and miR-875 in the prefrontal cortex. The interactions of ion channels, miRNAs, and disruptions of neurotransmitter networks can play an important role in the pathophysiology of MDD. Moreover, as shown in this study, ion channel inhibitors have significant potential in the treatment of this disease.


Asunto(s)
Encéfalo/efectos de los fármacos , Depresión/tratamiento farmacológico , Canales Iónicos/efectos de los fármacos , MicroARNs/efectos de los fármacos , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Depresión/metabolismo , Modelos Animales de Enfermedad , Masculino , MicroARNs/metabolismo , Ratas , Ratas Wistar , Estrés Psicológico/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPM/metabolismo
10.
Cell Calcium ; 101: 102521, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34953296

RESUMEN

TRPM7 is a non-selective divalent cation channel with an alpha-kinase domain. Corresponding with its broad expression, TRPM7 has a role in a wide range of cell functions, including proliferation, migration, and survival. Growing evidence shows that TRPM7 is also aberrantly expressed in various cancers, including brain cancers. Because ion channels have widespread tissue distribution and result in extensive physiological consequences when dysfunctional, these proteins can be compelling drug targets. In fact, ion channels comprise the third-largest drug target type, following enzymes and receptors. Literature has shown that suppression of TRPM7 results in inhibition of migration, invasion, and proliferation in several human brain tumours. Therefore, TRPM7 presents a potential target for therapeutic brain tumour interventions. This article reviews current literature on TRPM7 as a potential drug target in the context of brain tumours and provides an overview of various selective and non-selective modulators of the channel relevant to pharmacology, oncology, and ion channel function.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas , Canales Catiónicos TRPM , Neoplasias Encefálicas/tratamiento farmacológico , Humanos , Proteínas Serina-Treonina Quinasas , Canales Catiónicos TRPM/antagonistas & inhibidores
11.
Can J Physiol Pharmacol ; 100(2): 134-141, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34559972

RESUMEN

Fingolimod (FTY720) inhibits Ca2+-permeable, Mg2+-sensitive channels called transient receptor potential melastatin 7 (TRPM7), but its effects on Ca2+ paradox (CP) - induced myocardial damage has not been evaluated. We studied the effect of FTY720 on CP-induced myocardial damage and used other TRPM7 channel inhibitors nordihydroguaiaretic acid (NDGA) and Mg2+ to test if any effect of FTY720 was via TRPM7 inhibition. Langendorff-perfused Wistar rat hearts were treated with FTY720 or NDGA and subjected to a CP protocol consisting of Ca2+ depletion followed by Ca2+ repletion. Hearts of rats pre-treated with MgSO4 were also subjected to CP. Hemodynamic parameters were measured using an intraventricular balloon, and myocardial infarct size was quantified using triphenyltetrazolium chloride stain. TRPM7 proteins in ventricular tissue were detected using immunoblot analysis. FTY720, but not NDGA, decreased CP-induced infarct size. Both FTY720 and NDGA minimized the CP-induced elevation of left ventricular end-diastolic pressure, but only FTY720 ultimately improved ventricular developed pressure. Mg2+ pre-treatment had no effect on CP-induced infarct size, nor hemodynamic parameters during CP, nor the level of TRPM7 protein expression in ventricular tissue. Overall, FTY720 attenuated CP-induced myocardial damage, with potential therapeutic implications on Ca2+-mediated cardiotoxicity; however, the cardioprotective mechanism of FTY720 seems to be unrelated to TRPM7 channel modulation.


Asunto(s)
Calcio/efectos adversos , Calcio/metabolismo , Cardiotónicos , Clorhidrato de Fingolimod/farmacología , Infarto del Miocardio/tratamiento farmacológico , Animales , Clorhidrato de Fingolimod/uso terapéutico , Técnicas In Vitro , Magnesio/metabolismo , Masculino , Masoprocol/farmacología , Masoprocol/uso terapéutico , Infarto del Miocardio/etiología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Ratas Wistar , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo
12.
Sci Rep ; 11(1): 23232, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34853378

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Canales Catiónicos TRPM/agonistas , Canales Catiónicos TRPM/antagonistas & inhibidores , Andrógenos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Masculino , Invasividad Neoplásica , Receptores Androgénicos , Esferoides Celulares
13.
Nat Commun ; 12(1): 6815, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819493

RESUMEN

Bistable motoneurons of the spinal cord exhibit warmth-activated plateau potential driven by Na+ and triggered by a brief excitation. The thermoregulating molecular mechanisms of bistability and their role in motor functions remain unknown. Here, we identify thermosensitive Na+-permeable Trpm5 channels as the main molecular players for bistability in mouse motoneurons. Pharmacological, genetic or computational inhibition of Trpm5 occlude bistable-related properties (slow afterdepolarization, windup, plateau potentials) and reduce spinal locomotor outputs while central pattern generators for locomotion operate normally. At cellular level, Trpm5 is activated by a ryanodine-mediated Ca2+ release and turned off by Ca2+ reuptake through the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump. Mice in which Trpm5 is genetically silenced in most lumbar motoneurons develop hindlimb paresis and show difficulties in executing high-demanding locomotor tasks. Overall, by encoding bistability in motoneurons, Trpm5 appears indispensable for producing a postural tone in hindlimbs and amplifying the locomotor output.


Asunto(s)
Locomoción/fisiología , Neuronas Motoras/metabolismo , Paresia/fisiopatología , Médula Espinal/fisiología , Canales Catiónicos TRPM/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Simulación por Computador , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Células HEK293 , Miembro Posterior/fisiología , Humanos , Locomoción/efectos de los fármacos , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Paresia/genética , Técnicas de Placa-Clamp , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Médula Espinal/citología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética
14.
J Med Chem ; 64(22): 16820-16837, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34762442

RESUMEN

Transient receptor potential melastatin 8 (TRPM8) is crucially involved in pain modulation and perception, and TRPM8 antagonists have been proposed as potential therapeutic approaches for pain treatment. Previously, we developed two TRPM8 antagonists and proposed them as drug candidates for topical and systemic pain treatment. Here, we describe the design and synthesis of these two TRPM8 antagonists (27 and 45) and the rational approach of modulation/replacement of bioisosteric chemical groups, which allowed us to identify a combination of narrow ranges of pKa and LogD values that were crucial to ultimately optimize their potency and metabolic stability. Following the same approach, we then pursued the development of new TRPM8 antagonists suitable for the topical treatment of ocular painful conditions and identified two new compounds (51 and 59), N-alkoxy amide derivatives, that can permeate across ocular tissue and reduce the behavioral responses induced by the topical ocular menthol challenge in vivo.


Asunto(s)
Analgésicos/química , Analgésicos/farmacología , Descubrimiento de Drogas , Oftalmopatías/tratamiento farmacológico , Manejo del Dolor/métodos , Canales Catiónicos TRPM/antagonistas & inhibidores , Células HEK293 , Humanos , Relación Estructura-Actividad
15.
Bioorg Med Chem Lett ; 52: 128392, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34606996

RESUMEN

The preliminary results on the development of a viable methodology for the further functionalization of 4-hydroxythiazole derivatives to afford target TRPM8 antagonists are reported. The combined Sonogashira coupling/annulation reactions of the ethyl 2-(3-fluorophenyl)-4-tifluoromethylsulfonyloxy-1,3-thiazole-5-carboxylate have been applied to the synthesis of analogues of the selective blocker of TRPM8 DFL23448. Among all the synthetised derivatives, the most promising compound resulted to be active as TRPM8 blocker (IC50 = 4.06 µM), showing an excellent metabolic stability and no cytotoxic effects. Finally, in silico characterisation of the derivatives showed no violation of the drug-likeness rules.


Asunto(s)
Diseño de Fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Tiazoles/farmacología , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Estructura Molecular , Relación Estructura-Actividad , Canales Catiónicos TRPM/metabolismo , Tiazoles/síntesis química , Tiazoles/química
16.
Neural Plast ; 2021: 8774663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659399

RESUMEN

Hippocampal cell death and cognitive dysfunction are common following global cerebral ischemia across all ages, including children. Most research has focused on preventing neuronal death. Restoration of neuronal function after cell death is an alternative approach (neurorestoration). We previously identified transient receptor potential M2 (TRPM2) ion channels as a potential target for acute neuroprotection and delayed neurorestoration in an adult CA/CPR mouse model. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in juvenile (p20-25) mice was used to investigate the role of ion TRPM2 channels in neuroprotection and ischemia-induced synaptic dysfunction in the developing brain. Our novel TRPM2 inhibitor, tatM2NX, did not confer protection against CA1 pyramidal cell death but attenuated synaptic plasticity (long-term plasticity (LTP)) deficits in both sexes. Further, in vivo administration of tatM2NX two weeks after CA/CPR reduced LTP impairments and restored memory function. These data provide evidence that pharmacological synaptic restoration of the surviving hippocampal network can occur independent of neuroprotection via inhibition of TRPM2 channels, providing a novel strategy to improve cognitive recovery in children following cerebral ischemia. Importantly, these data underscore the importance of age-appropriate models in disease research.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Recuperación de la Función/fisiología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/fisiología , Factores de Edad , Animales , Isquemia Encefálica/fisiopatología , Reanimación Cardiopulmonar/métodos , Femenino , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Técnicas de Cultivo de Órganos , Recuperación de la Función/efectos de los fármacos
17.
Adv Sci (Weinh) ; 8(22): e2101717, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34658162

RESUMEN

Platinum-based compounds in chemotherapy such as oxaliplatin often induce peripheral neuropathy and neuropathic pain such as cold allodynia in patients. Transient Receptor Potential Melastatin 8 (TRPM8) ion channel is a nociceptor critically involved in such pathological processes. Direct blockade of TRPM8 exhibits significant analgesic effects but also incurs severe side effects such as hypothermia. To selectively target TRPM8 channels against cold allodynia, a cyclic peptide DeC-1.2 is de novo designed with the optimized hot-spot centric approach. DeC-1.2 modality specifically inhibited the ligand activation of TRPM8 but not the cold activation as measured in single-channel patch clamp recordings. It is further demonstrated that DeC-1.2 abolishes cold allodynia in oxaliplatin treated mice without altering body temperature, indicating DeC-1.2 has the potential for further development as a novel analgesic against oxaliplatin-induced neuropathic pain.


Asunto(s)
Antineoplásicos/efectos adversos , Hiperalgesia/prevención & control , Oxaliplatino/efectos adversos , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética , Animales , Antineoplásicos/metabolismo , Frío , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Ratones , Oxaliplatino/metabolismo , Canales Catiónicos TRPM/metabolismo
18.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502410

RESUMEN

Transient receptor potential melastatin 4 (TRPM4) plays an important role in many tissues, including pacemaker and conductive tissues of the heart, but much less is known about its electrophysiological role in ventricular myocytes. Our earlier results showed the lack of selectivity of 9-phenanthrol, so CBA ((4-chloro-2-(2-chlorophenoxy)acetamido) benzoic acid) was chosen as a new, potentially selective inhibitor. Goal: Our aim was to elucidate the effect and selectivity of CBA in canine left ventricular cardiomyocytes and to study the expression of TRPM4 in the canine heart. Experiments were carried out in enzymatically isolated canine left ventricular cardiomyocytes. Ionic currents were recorded with an action potential (AP) voltage-clamp technique in whole-cell configuration at 37 °C. An amount of 10 mM BAPTA was used in the pipette solution to exclude the potential activation of TRPM4 channels. AP was recorded with conventional sharp microelectrodes. CBA was used in 10 µM concentrations. Expression of TRPM4 protein in the heart was studied by Western blot. TRPM4 protein was expressed in the wall of all four chambers of the canine heart as well as in samples prepared from isolated left ventricular cells. CBA induced an approximately 9% reduction in AP duration measured at 75% and 90% of repolarization and decreased the short-term variability of APD90. Moreover, AP amplitude was increased and the maximal rates of phase 0 and 1 were reduced by the drug. In AP clamp measurements, CBA-sensitive current contained a short, early outward and mainly a long, inward current. Transient outward potassium current (Ito) and late sodium current (INa,L) were reduced by approximately 20% and 47%, respectively, in the presence of CBA, while L-type calcium and inward rectifier potassium currents were not affected. These effects of CBA were largely reversible upon washout. Based on our results, the CBA induced reduction of phase-1 slope and the slight increase of AP amplitude could have been due to the inhibition of Ito. The tendency for AP shortening can be explained by the inhibition of inward currents seen in AP-clamp recordings during the plateau phase. This inward current reduced by CBA is possibly INa,L, therefore, CBA is not entirely selective for TRPM4 channels. As a consequence, similarly to 9-phenanthrol, it cannot be used to test the contribution of TRPM4 channels to cardiac electrophysiology in ventricular cells, or at least caution must be applied.


Asunto(s)
Canales Catiónicos TRPM/metabolismo , Función Ventricular/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Ácido Benzoico/farmacología , Calcio/metabolismo , Electrofisiología Cardíaca , Perros , Fenómenos Electrofisiológicos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/patología , Masculino , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Sodio/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/fisiología
19.
Mol Neurobiol ; 58(11): 5581-5601, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34370177

RESUMEN

Excessive Ca2+ influx and mitochondrial oxidative stress (OS) of trigeminal ganglia (TG) have essential roles in the etiology of migraine headache and aura. The stimulation of TRPM2 channel via the generation of OS and ADP-ribose (ADPR) induces pain, inflammatory, and oxidative neurotoxicity, although its inhibition reduces the intensity of pain and neurotoxicity in several neurons. However, the cellular and molecular effects of TRPM2 in the TG of migraine model (glyceryl trinitrate, GTN) on the induction of pain, OS, apoptosis, and inflammation remain elusive. GTN-mediated increases of pain intensity, apoptosis, death, cytosolic reactive oxygen species (ROS), mitochondrial ROS, caspase -3, caspase -9, cytosolic Ca2+ levels, and cytokine generations (TNF-α, IL-1ß, and IL-6) in the TG of TRPM2 wild-type mouse were further increased by the TRPM2 activation, although they were modulated by the treatments of GSH, PARP-1 inhibitors (PJ34 and DPQ), and TRPM2 blockers (ACA and 2APB). However, the effects of GTN were not observed in the TG of TRPM2 knockout mice. The current data indicate that the maintaining activation of TRPM2 is not only important for the quenching OS, inflammation, and neurotoxicity in the TG neurons of mice with experimental migraine but also equally critical to the modulation of GTN-induced pain.


Asunto(s)
Trastornos Migrañosos/metabolismo , Canales Catiónicos TRPM/fisiología , Ganglio del Trigémino/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Apoptosis , Compuestos de Boro/farmacología , Señalización del Calcio , Caspasas/metabolismo , Cinamatos/farmacología , Citocinas/biosíntesis , Citocinas/genética , Activación Enzimática , Glutatión/metabolismo , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Peroxidación de Lípido , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/fisiopatología , Enfermedades Neuroinflamatorias , Neuronas/patología , Nitroglicerina/toxicidad , Estrés Oxidativo , Fenantrenos/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/deficiencia , Ganglio del Trigémino/efectos de los fármacos , ortoaminobenzoatos/farmacología
20.
Eur J Med Chem ; 225: 113750, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34416664

RESUMEN

Transient receptor potential melastatin 2 (TRPM2) channel is associated with ischemia/reperfusion injury, inflammation, cancer and neurodegenerative diseases. However, the lack of specific inhibitors impedes the development of TRPM2 targeted therapeutic agents. To develop a selective TRPM2 inhibitor, three-dimensional similarity-based screening strategy was employed using the energy-minimized conformation of non-selective TRPM2 inhibitor 2-APB as the query structure, which resulted in the discovery of a novel tricyclic TRPM2 inhibitor Z-4 with benzo[d]imidazo[1,2-a]imidazole skeleton. A series of Z-4 derivatives were subsequently synthesized and evaluated using calcium imaging and electrophysiology approaches. Among them, preferred compounds ZA10 and ZA18 inhibited the TRPM2 channel with micromolar half-maximal inhibitory concentration values and exhibited TRPM2 selectivity over the TRPM8 channel, TRPV1 channel, InsP3 receptor and Orai channel. The analysis of structure-activity relationship provides valuable insights for further development of selective TRPM2 inhibitors. Neuroprotection assay showed that ZA10 and ZA18 could effectively reduce the mortality of SH-SY5Y cells induced by H2O2. These findings enrich the structure types of existing TRPM2 inhibitors and might provide a new tool for the study of TRPM2 function in Reactive oxygen species (ROS) -related diseases.


Asunto(s)
Diseño de Fármacos , Imidazoles/farmacología , Fármacos Neuroprotectores/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Imidazoles/síntesis química , Imidazoles/química , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Relación Estructura-Actividad , Canales Catiónicos TRPM/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...