Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 414
Filtrar
1.
Eur J Pharmacol ; 953: 175833, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37290679

RESUMEN

Borneol has been used successfully for the treatment of itchy skin in traditional Chinese medicine. However, the antipruritic effect of borneol has rarely been studied, and the mechanism is unclear. Here, we showed that topical application of borneol on skin substantially suppressed pruritogen chloroquine- and compound 48/80-induced itching in mice. The potential targets of borneol, including transient receptor potential cation channel subfamily V member 3 (TRPV3), transient receptor potential cation channel subfamily A member 1 (TRPA1), transient receptor potential cation channel subfamily M member 8 (TRPM8), and gamma-aminobutyric acid type A (GABAA) receptor were pharmacologically inhibited or genetically knocked out one by one in mouse. Itching behavior studies demonstrated that the antipruritic effect of borneol is largely independent of TRPV3 and GABAA receptor, and TRPA1 and TRPM8 channels are responsible for a major portion of the effect of borneol on chloroquine-induced nonhistaminergic itching. Borneol activates TRPM8 and inhibits TRPA1 in sensory neurons of mice. Topical co-application of TRPA1 antagonist and TRPM8 agonist mimicked the effect of borneol on chloroquine-induced itching. Intrathecal injection of a group II metabotropic glutamate receptor antagonist partially attenuated the effect of borneol and completely abolished the effect of TRPM8 agonist on chloroquine-induced itching, suggesting that a spinal glutamatergic mechanism is involved. In contrast, the effect of borneol on compound 48/80-induced histaminergic itching occurs through TRPA1-and TRPM8-independent mechanisms. Our work demonstrates that borneol is an effective topical itch reliever, and TRPA1 inhibition and TRPM8 activation in peripheral nerve terminals account for its antipruritic effect.


Asunto(s)
Canales Catiónicos TRPM , Canales de Potencial de Receptor Transitorio , Ratones , Animales , Antipruriginosos/farmacología , Antipruriginosos/uso terapéutico , Canal Catiónico TRPA1 , Canales Catiónicos TRPM/fisiología , Prurito/inducido químicamente , Prurito/tratamiento farmacológico , Células Receptoras Sensoriales , Cloroquina/farmacología , Nervios Periféricos , Canales Catiónicos TRPV
2.
Curr Probl Cardiol ; 48(2): 101012, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34644560

RESUMEN

Transient receptor potential (TRP) family play critical roles in cardiovascular system. TRPM family as largest TRP subfamily is non-voltage Ca2+-activated selective channels which has 8 members. This study aimed to discuss the role of TRPM family in cardiovascular system and diseases. Systematic search was performed covering PubMed, ISI Web of Science, and Google Scholar from inception until June 2021 using related keywords and Mesh terms for English studies with human, animal and in-vitro subjects. Finally 10 studies were selected for data extraction. Reviewing the articles showed that TRPM2, TRPM4, TRPM5, TRPM6 and TRPM7 play important roles in cardiovascular system and diseases. TRPM2 could be activated by reactive oxygen species (ROS) and effects on cardiac injury and cardiac fibrosis. TRPM7 and TRPM6 also have been reported to be associated with cardiac fibrosis and atrial fibrosis development respectively. TRPM4 channels contributed to resting membrane potential of cerebral artery smooth muscle cells and atrial contraction. TRPM5 channels are bitter taste sensors and prevent high salt intake and consequently high blood pressure due to the high salt intake. In conclusion based on the proof of the effectiveness of some members of TRPM family in the cardiovascular system, research on other members of this channel group seems to be useful and necessary to find their possible connection to the cardiovascular system.


Asunto(s)
Sistema Cardiovascular , Canales Catiónicos TRPM , Animales , Humanos , Canales Catiónicos TRPM/fisiología , Cloruro de Sodio Dietético , Potenciales de la Membrana , Clusterina , Proteínas Serina-Treonina Quinasas
3.
Rev Med Chil ; 150(1): 88-92, 2022 Jan.
Artículo en Español | MEDLINE | ID: mdl-35856969

RESUMEN

D. Julius was awarded the 2021 Medicine Nobel prize for the discovery of new cationic channels that detect temperatures either over 40 °C (TRPV1) or cold (TRPM8) ranging from 8-15 °C, followed by the latter identification of other channels that sense temperatures within other ranges. On the other hand, A. Patapoutian shared the 2021 Nobel prize for the independent and simultaneous co-discovery of the TRPM8 cationic channel. Furthermore, Patapoutian iden-tified piezo 1 and 2 channels previously referred to as the cell mechanosensors related to the sense of touch and proprioception. These experimental findings indicate that these novel cationic channels localized in nerve endings of the skin, mouth, lips, bronchial tree, the nephron, plus a variety of tissues transduce phy-sical stimuli into electrical activity that reach the brain sensory cortex to process these stimuli and elicit animal behavior.


Asunto(s)
Canales Catiónicos TRPM , Animales , Frío , Premio Nobel , Piel , Canales Catiónicos TRPM/fisiología , Temperatura
4.
J Smooth Muscle Res ; 58(0): 11-21, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35354708

RESUMEN

In the lower urinary tract, transient receptor potential (TRP) channels are primarily involved in physiological function, especially in cellular sensors responding to chemical and physical stimuli. Among TRP channels, TRP melastatin 8 (TRPM8) channels, responding to cold temperature and/or chemical agents, such as menthol or icilin, are mainly expressed in the nerve endings of the primary afferent neurons and in the cell bodies of dorsal root ganglia innervating the urinary bladder (via Aδ- and C-fibers); this suggests that TRPM8 channels primarily contribute to bladder sensory (afferent) function. Storage symptoms of overactive bladder, benign prostatic hyperplasia, and interstitial cystitis are commonly related to sensory function (bladder hypersensitivity); thus, TRPM8 channels may also contribute to the pathophysiology of bladder hypersensitivity. Indeed, it has been reported in a pharmacological investigation using rodents that TRPM8 channels contribute to the pathophysiological bladder afferent hypersensitivity of mechanosensitive C-fibers. Similar findings have also been reported in humans. Therefore, a TRPM8 antagonist would be a promising therapeutic target for bladder hypersensitive disorders, including urinary urgency or nociceptive pain. In this review article, the functional role of the TRPM8 channel in the lower urinary tract and the potential of its antagonist for the treatment of bladder disorders was described.


Asunto(s)
Canales Catiónicos TRPM , Enfermedades de la Vejiga Urinaria , Ganglios Espinales , Humanos , Proteínas de la Membrana , Mentol/farmacología , Mentol/uso terapéutico , Canales Catiónicos TRPM/fisiología , Vejiga Urinaria , Enfermedades de la Vejiga Urinaria/tratamiento farmacológico
5.
Rev. méd. Chile ; 150(1): 88-92, ene. 2022. ilus
Artículo en Español | LILACS | ID: biblio-1389622

RESUMEN

D. Julius was awarded the 2021 Medicine Nobel prize for the discovery of new cationic channels that detect temperatures either over 40 °C (TRPV1) or cold (TRPM8) ranging from 8-15 °C, followed by the latter identification of other channels that sense temperatures within other ranges. On the other hand, A. Patapoutian shared the 2021 Nobel prize for the independent and simultaneous co-discovery of the TRPM8 cationic channel. Furthermore, Patapoutian iden-tified piezo 1 and 2 channels previously referred to as the cell mechanosensors related to the sense of touch and proprioception. These experimental findings indicate that these novel cationic channels localized in nerve endings of the skin, mouth, lips, bronchial tree, the nephron, plus a variety of tissues transduce phy-sical stimuli into electrical activity that reach the brain sensory cortex to process these stimuli and elicit animal behavior.


Asunto(s)
Animales , Canales Catiónicos TRPM/fisiología , Piel , Temperatura , Frío , Premio Nobel
6.
Acta Pharmacol Sin ; 43(4): 759-770, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34108651

RESUMEN

Ion channels are ubiquitously expressed in almost all living cells, and are the third-largest category of drug targets, following enzymes and receptors. The transient receptor potential melastatin (TRPM) subfamily of ion channels are important to cell function and survival. Studies have shown upregulation of the TRPM family of ion channels in various brain tumours. Gliomas are the most prevalent form of primary malignant brain tumours with no effective treatment; thus, drug development is eagerly needed. TRPM2 is an essential ion channel for cell function and has important roles in oxidative stress and inflammation. In response to oxidative stress, ADP-ribose (ADPR) is produced, and in turn activates TRPM2 by binding to the NUDT9-H domain on the C-terminal. TRPM2 has been implicated in various cancers and is significantly upregulated in brain tumours. This article reviews the current understanding of TRPM2 in the context of brain tumours and overviews the effects of potential drug therapies targeting TRPM2 including hydrogen peroxide (H2O2), curcumin, docetaxel and selenium, paclitaxel and resveratrol, and botulinum toxin. It is long withstanding knowledge that gliomas are difficult to treat effectively, therefore investigating TRPM2 as a potential therapeutic target for brain tumours may be of considerable interest in the fields of ion channels and pharmacology.


Asunto(s)
Neoplasias Encefálicas , Canales Catiónicos TRPM , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Adenosina Difosfato Ribosa/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Calcio/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo , Canales Catiónicos TRPM/fisiología
7.
PLoS Biol ; 19(12): e3001496, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34928937

RESUMEN

Magnesium is essential for cellular life, but how it is homeostatically controlled still remains poorly understood. Here, we report that members of CNNM family, which have been controversially implicated in both cellular Mg2+ influx and efflux, selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. Coexpression of CNNMs with the channel markedly increased uptake of divalent cations, which is prevented by an inactivating mutation to the channel's pore. Knockout (KO) of TRPM7 in cells or application of the TRPM7 channel inhibitor NS8593 also interfered with CNNM-stimulated divalent cation uptake. Conversely, KO of CNNM3 and CNNM4 in HEK-293 cells significantly reduced TRPM7-mediated divalent cation entry, without affecting TRPM7 protein expression or its cell surface levels. Furthermore, we found that cellular overexpression of phosphatases of regenerating liver (PRLs), known CNNMs binding partners, stimulated TRPM7-dependent divalent cation entry and that CNNMs were required for this activity. Whole-cell electrophysiological recordings demonstrated that deletion of CNNM3 and CNNM4 from HEK-293 cells interfered with heterologously expressed and native TRPM7 channel function. We conclude that CNNMs employ the TRPM7 channel to mediate divalent cation influx and that CNNMs also possess separate TRPM7-independent Mg2+ efflux activities that contribute to CNNMs' control of cellular Mg2+ homeostasis.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Ciclinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPM/metabolismo , Proteínas de Transporte de Catión/fisiología , Cationes Bivalentes/metabolismo , Línea Celular Tumoral , Ciclinas/fisiología , Células HEK293 , Humanos , Magnesio/metabolismo , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/fisiología , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/fisiología
8.
Cell Rep ; 37(7): 110025, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34788616

RESUMEN

Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable cation channel, is gated by intracellular adenosine diphosphate ribose (ADPR), Ca2+, warm temperature, and oxidative stress. It is critically involved in physiological and pathological processes ranging from inflammation to stroke to neurodegeneration. At present, the channel's gating and ion permeation mechanisms, such as the location and identity of the selectivity filter, remain ambiguous. Here, we report the cryo-electron microscopy (cryo-EM) structure of human TRPM2 in nanodisc in the ligand-free state. Cryo-EM map-guided computational modeling and patch-clamp recording further identify a quadruple-residue motif as the ion selectivity filter, which adopts a restrictive conformation in the closed state and acts as a gate, profoundly contrasting with its widely open conformation in the Nematostella vectensis TRPM2. Our study reveals the gating of human TRPM2 by the filter and demonstrates the feasibility of using cryo-EM in conjunction with computational modeling and functional studies to garner structural information for intrinsically dynamic but functionally important domains.


Asunto(s)
Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/fisiología , Sitios de Unión/fisiología , Calcio/metabolismo , Cationes , Microscopía por Crioelectrón/métodos , Humanos , Activación del Canal Iónico/fisiología , Técnicas de Placa-Clamp/métodos , Unión Proteica/fisiología , Canales Catiónicos TRPM/ultraestructura
9.
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
10.
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
11.
Oxid Med Cell Longev ; 2021: 7356266, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367466

RESUMEN

Cerebral ischemia-reperfusion (I-R) transiently increased autophagy by producing excessively reactive oxygen species (ROS); on the other hand, activated autophagy would remove ROS-damaged mitochondria and proteins, which led to cell survival. However, the regulation mechanism of autophagy activity during cerebral I-R is still unclear. In this study, we found that deficiency of the TRPM2 channel which is a ROS sensor significantly decreased I-R-induced neuronal damage. I-R transiently increased autophagy activity both in vitro and in vivo. More importantly, TRPM2 deficiency decreased I-R-induced neurological deficit score and infarct volume. Interestingly, our results indicated that TRPM2 deficiency could further activate AMPK rather than Beclin1 activity, suggesting that TRPM2 inhibits autophagy by regulating the AMPK/mTOR pathway in I-R. In conclusion, our study reveals that ROS-activated TRPM2 inhibits autophagy by downregulating the AMPK/mTOR pathway, which results in neuronal death induced by cerebral I-R, further supporting that TRPM2 might be a potential drug target for cerebral ischemic injury therapy.


Asunto(s)
Autofagia , Isquemia Encefálica/prevención & control , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/prevención & control , Canales Catiónicos TRPM/fisiología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Estrés Oxidativo , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
12.
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
13.
Int J Mol Sci ; 22(16)2021 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-34445449

RESUMEN

The cardiac Mg2+-sensitive, TRPM6, and TRPM7-like channels remain undefined, especially with the uncertainty regarding TRPM6 expression in cardiomyocytes. Additionally, their contribution to the cardiac action potential (AP) profile is unclear. Immunofluorescence assays showed the expression of the TRPM6 and TRPM7 proteins in isolated pig atrial and ventricular cardiomyocytes, of which the expression was modulated by incubation in extracellular divalent cation-free conditions. In patch clamp studies of cells dialyzed with solutions containing zero intracellular Mg2+ concentration ([Mg2+]i) to activate the Mg2+-sensitive channels, raising extracellular [Mg2+] ([Mg2+]o) from the 0.9-mM baseline to 7.2 mM prolonged the AP duration (APD). In contrast, no such effect was observed in cells dialyzed with physiological [Mg2+]i. Under voltage clamp, in cells dialyzed with zero [Mg2+]i, depolarizing ramps induced an outward-rectifying current, which was suppressed by raising [Mg2+]o and was absent in cells dialyzed with physiological [Mg2+]i. In cells dialyzed with physiological [Mg2+]i, raising [Mg2+]o decreased the L-type Ca2+ current and the total delayed-rectifier current but had no effect on the APD. These results suggest a co-expression of the TRPM6 and TRPM7 proteins in cardiomyocytes, which are therefore the molecular candidates for the native cardiac Mg2+-sensitive channels, and also suggest that the cardiac Mg2+-sensitive current shortens the APD, with potential implications in arrhythmogenesis.


Asunto(s)
Potenciales de Acción , Magnesio/metabolismo , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Cationes Bivalentes , Miocitos Cardíacos/fisiología , Sus scrofa/metabolismo , Sus scrofa/fisiología , Canales Catiónicos TRPM/fisiología
14.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34299254

RESUMEN

The transient receptor potential (TRP) melastatin-like subfamily member 2 (TRPM2) is a non-selective calcium-permeable cation channel. It is expressed by many mammalian tissues, including bone marrow, spleen, lungs, heart, liver, neutrophils, and endothelial cells. The best-known mechanism of TRPM2 activation is related to the binding of ADP-ribose to the nudix-box sequence motif (NUDT9-H) in the C-terminal domain of the channel. In cells, the production of ADP-ribose is a result of increased oxidative stress. In the context of endothelial function, TRPM2-dependent calcium influx seems to be particularly interesting as it participates in the regulation of barrier function, cell death, cell migration, and angiogenesis. Any impairments of these functions may result in endothelial dysfunction observed in such conditions as atherosclerosis or hypertension. Thus, TRPM2 seems to be an attractive therapeutic target for the conditions connected with the increased production of reactive oxygen species. However, before the application of TRPM2 inhibitors will be possible, some issues need to be resolved. The main issues are the lack of specificity, poor membrane permeabilization, and low stability in in vivo conditions. The article aims to summarize the latest findings on a role of TRPM2 in endothelial cells. We also show some future perspectives for the application of TRPM2 inhibitors in cardiovascular system diseases.


Asunto(s)
Células Endoteliales/metabolismo , Canales Catiónicos TRPM/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Calcio/metabolismo , Muerte Celular , Movimiento Celular , Células Endoteliales/fisiología , Humanos , Activación del Canal Iónico/fisiología , Estrés Oxidativo/fisiología , Pirofosfatasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/fisiología
15.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067824

RESUMEN

Pyramidal neurons in the medial prefrontal cortical layer 2/3 are an essential contributor to the cellular basis of working memory; thus, changes in their intrinsic excitability critically affect medial prefrontal cortex (mPFC) functional properties. Transient Receptor Potential Melastatin 4 (TRPM4), a calcium-activated nonselective cation channel (CAN), regulates the membrane potential in a calcium-dependent manner. In this study, we uncovered the role of TRPM4 in regulating the intrinsic excitability plasticity of pyramidal neurons in the mouse mPFC layer of 2/3 using a combination of conventional and nystatin perforated whole-cell recordings. Interestingly, we found that TRPM4 is open at resting membrane potential, and its inhibition increases input resistance and hyperpolarizes membrane potential. After high-frequency stimulation, pyramidal neurons increase a calcium-activated non-selective cation current, increase the action potential firing, and the amplitude of the afterdepolarization, these effects depend on intracellular calcium. Furthermore, pharmacological inhibition or genetic silencing of TRPM4 reduces the firing rate and the afterdepolarization after high frequency stimulation. Together, these results show that TRPM4 plays a significant role in the excitability of mPFC layer 2/3 pyramidal neurons by modulating neuronal excitability in a calcium-dependent manner.


Asunto(s)
Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Canales Catiónicos TRPM/metabolismo , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Corteza Prefrontal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Canales Catiónicos TRPM/fisiología
16.
J Am Soc Nephrol ; 32(9): 2125-2136, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34045316

RESUMEN

Magnesium is an essential cofactor in many cellular processes, and aberrations in magnesium homeostasis can have life-threatening consequences. The kidney plays a central role in maintaining serum magnesium within a narrow range (0.70-1.10 mmol/L). Along the proximal tubule and thick ascending limb, magnesium reabsorption occurs via paracellular pathways. Members of the claudin family form the magnesium pores in these segments, and also regulate magnesium reabsorption by adjusting the transepithelial voltage that drives it. Along the distal convoluted tubule transcellular reabsorption via heteromeric TRPM6/7 channels predominates, although paracellular reabsorption may also occur. In this segment, the NaCl cotransporter plays a critical role in determining transcellular magnesium reabsorption. Although the general machinery involved in renal magnesium reabsorption has been identified by studying genetic forms of magnesium imbalance, the mechanisms regulating it are poorly understood. This review discusses pathways of renal magnesium reabsorption by different segments of the nephron, emphasizing newer findings that provide insight into regulatory process, and outlining critical unanswered questions.


Asunto(s)
Magnesio/metabolismo , Reabsorción Renal/fisiología , Claudinas/fisiología , Humanos , Nefronas/fisiopatología , Proteínas Serina-Treonina Quinasas/fisiología , Canales Catiónicos TRPM/fisiología
17.
Elife ; 102021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34032213

RESUMEN

Upon exposure to harmful microorganisms, hosts engage in protective molecular and behavioral immune responses, both of which are ultimately regulated by the nervous system. Using the nematode Caenorhabditis elegans, we show that ingestion of Enterococcus faecalis leads to a fast pathogen avoidance behavior that results in aversive learning. We have identified multiple sensory mechanisms involved in the regulation of avoidance of E. faecalis. The G-protein coupled receptor NPR-1-dependent oxygen-sensing pathway opposes this avoidance behavior, while an ASE neuron-dependent pathway and an AWB and AWC neuron-dependent pathway are directly required for avoidance. Colonization of the anterior part of the intestine by E. faecalis leads to AWB and AWC mediated olfactory aversive learning. Finally, two transient receptor potential melastatin (TRPM) channels, GON-2 and GTL-2, mediate this newly described rapid pathogen avoidance. These results suggest a mechanism by which TRPM channels may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance and aversive learning by detecting changes in host physiology.


Asunto(s)
Reacción de Prevención , Caenorhabditis elegans/microbiología , Enterococcus faecalis/patogenicidad , Intestinos/microbiología , Canales Catiónicos TRPM/fisiología , Animales , Enterococcus faecalis/aislamiento & purificación , Interacciones Huésped-Patógeno , Intestinos/patología , Neuronas/metabolismo , Virulencia
18.
Sci Rep ; 11(1): 9339, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33927223

RESUMEN

Neutrophils must navigate accurately towards pathogens in order to destroy invaders and thus defend our bodies against infection. Here we show that hydrogen peroxide, a potent neutrophil chemoattractant, guides chemotaxis by activating calcium-permeable TRPM2 ion channels and generating an intracellular leading-edge calcium "pulse". The thermal sensitivity of TRPM2 activation means that chemotaxis towards hydrogen peroxide is strongly promoted by small temperature elevations, suggesting that an important function of fever may be to enhance neutrophil chemotaxis by facilitating calcium influx through TRPM2. Chemotaxis towards conventional chemoattractants such as LPS, CXCL2 and C5a does not depend on TRPM2 but is driven in a similar way by leading-edge calcium pulses. Other proposed initiators of neutrophil movement, such as PI3K, Rac and lyn, influence chemotaxis by modulating the amplitude of calcium pulses. We propose that intracellular leading-edge calcium pulses are universal drivers of the motile machinery involved in neutrophil chemotaxis.


Asunto(s)
Quimiotaxis , Neutrófilos/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Peróxido de Hidrógeno , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Transl Res ; 233: 127-143, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33691194

RESUMEN

Ophiocordyceps sinensis (OCS), an entomopathogenic fungus, is known to exert antiproliferative and antitissue remodeling effects. Vascular remodeling and vasoconstriction play critical roles in the development of pulmonary hypertension (PH). The therapeutic potential of OCS for PH was investigated using rodent PH models, and cultured pulmonary artery endothelial and smooth muscle cells (PAECs and PASMCs), with a focus on the involvement of TRPM7. OCS ameliorated the development of PH, right ventricular hypertrophy and dysfunction in the monocrotaline-induced PH rats. The genetic knockout of TRPM7 attenuated the development of PH in mice with monocrotaline pyrrole-induced PH. TRPM7 was associated with medial hypertrophy and the plexiform lesions in rats and humans with PH. OCS suppressed proliferation of PASMCs derived from the PH patients. Ethanol extracts of OCS inhibited TRPM7-like current, TGF-ß2-induced endothelial-mesenchymal transition, IL-6-induced STAT3 phosphorylation, and PDGF-induced Akt phosphorylation in PAECs or PASMCs. These inhibitory effects were recapitulated by either siRNA-mediated TRPM7 knockdown or treatment with TRPM7 antagonist FTY-720. OCS and FTY-720 induced vasorelaxation in the isolated normal human pulmonary artery. As a result, the present study proposes the therapeutic potential of OCS for the treatment of PH. The inhibition of TRPM7 is suggested to underlie the therapeutic effect of OCS.


Asunto(s)
Cordyceps/fisiología , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/terapia , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Hipertensión Pulmonar/patología , Masculino , Medicina Tradicional China , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Canales Catiónicos TRPM/deficiencia , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/fisiología , Investigación Biomédica Traslacional , Vasodilatación
20.
J Invest Dermatol ; 141(2): 250-252, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33504438

RESUMEN

Resistance to targeted therapy and immunotherapy remains a major obstacle in improving care for patients with advanced melanoma. MicroRNAs play important roles in regulating gene networks involved in disease progression and resistance to therapy in cancers such as melanoma. MicroRNA miR-211 contributes to melanocyte and melanoma biology and has been implicated in targeted therapy resistance. Lee et al. (2020) report a novel mechanism by which miR-211 promotes resistance to BRAFV600E inhibitor therapy via the upregulation of the extracellular signal-regulated kinase 5 signaling pathway.


Asunto(s)
Melanoma/tratamiento farmacológico , MicroARNs/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Resistencia a Antineoplásicos , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Canales Catiónicos TRPM/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...