Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(9)2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946275

RESUMEN

The tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b/PEX5R) is an interaction partner and auxiliary subunit of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which are key for rhythm generation in the brain and in the heart. Since TRIP8b is expressed in central neurons but not in cardiomyocytes, the TRIP8b-HCN interaction has been studied intensely in the brain, but is deemed irrelevant in the cardiac conduction system. Still, to date, TRIP8b has not been studied in the intrinsic cardiac nervous system (ICNS), a neuronal network located within epicardial fat pads. In vitro electrophysiological studies revealed that TRIP8b-deficient mouse hearts exhibit increased atrial refractory and atrioventricular nodal refractory periods, compared to hearts of wild-type littermates. Meanwhile, heart rate, sino-nodal recovery time, and ventricular refractory period did not differ between genotypes. Trip8b mRNA was detected in the ICNS by quantitative polymerase chain reaction. RNAscope in situ hybridization confirmed Trip8b localization in neuronal somata and nerve fibers. Additionally, we found a very low amount of mRNAs in the sinus node and atrioventricular node, most likely attributable to the delicate fibers innervating the conduction system. In contrast, TRIP8b protein was not detectable. Our data suggest that TRIP8b in the ICNS may play a role in the modulation of atrial electrophysiology beyond HCN-mediated sino-nodal control of the heart.


Asunto(s)
Corazón/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas de la Membrana/metabolismo , Peroxinas/metabolismo , Animales , Eliminación de Gen , Expresión Génica , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Peroxinas/genética , Mapas de Interacción de Proteínas , ARN Mensajero/genética
2.
Pflugers Arch ; 472(12): 1733-1742, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33095298

RESUMEN

The hyperpolarization-activated cation current If is a key determinant for cardiac pacemaker activity. It is conducted by subunits of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel family, of which HCN4 is predominant in mammalian heart. Both loss-of-function and gain-of-function mutations of the HCN4 gene are associated with sinus node dysfunction in humans; however, their functional impact is not fully understood yet. Here, we sought to characterize a HCN4 V759I variant detected in a patient with a family history of sick sinus syndrome. The genomic analysis yielded a mono-allelic HCN4 V759I variant in a 49-year-old woman presenting with a family history of sick sinus syndrome. This HCN4 variant was previously classified as putatively pathogenic because genetically linked to sudden infant death syndrome and malignant epilepsy. However, detailed electrophysiological and cell biological characterization of HCN4 V759I in Xenopus laevis oocytes and embryonic rat cardiomyocytes, respectively, did not reveal any obvious abnormality. Voltage dependence and kinetics of mutant channel activation, modulation of cAMP-gating by the neuronal HCN channel auxiliary subunit PEX5R, and cell surface expression were indistinguishable from wild-type HCN4. In good agreement, the clinically likewise affected mother of the patient does not exhibit the reported HCN4 variance. HCN4 V759I resembles an innocuous genetic HCN channel variant, which is not sufficient to disturb cardiac pacemaking. Once more, our work emphasizes the importance of careful functional interpretation of genetic findings not only in the context of hereditary cardiac arrhythmias.


Asunto(s)
Bradicardia/genética , Frecuencia Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Mutación Missense , Canales de Potasio/genética , Potenciales de Acción , Animales , Bradicardia/diagnóstico , Bradicardia/fisiopatología , Células Cultivadas , Femenino , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Persona de Mediana Edad , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Canales de Potasio/metabolismo , Transporte de Proteínas , Ratas , Ratas Wistar , Xenopus
3.
Nat Chem Biol ; 14(8): 764-767, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013061

RESUMEN

L-type Ca2+ channels (LTCCs) play a crucial role in excitation-contraction coupling and release of hormones from secretory cells. They are targets of antihypertensive and antiarrhythmic drugs such as diltiazem. Here, we present a photoswitchable diltiazem, FHU-779, which can be used to reversibly block endogenous LTCCs by light. FHU-779 is as potent as diltiazem and can be used to place pancreatic ß-cell function and cardiac activity under optical control.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Diltiazem/farmacología , Colorantes Fluorescentes/farmacología , Corazón/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Imagen Óptica , Canales de Calcio Tipo L/química , Diltiazem/química , Colorantes Fluorescentes/química , Humanos , Células Secretoras de Insulina/metabolismo , Luz , Procesos Fotoquímicos
4.
Angew Chem Int Ed Engl ; 58(43): 15421-15428, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31441199

RESUMEN

Photopharmacology relies on ligands that change their pharmacodynamics upon photoisomerization. Many of these ligands are azobenzenes that are thermodynamically more stable in their elongated trans-configuration. Often, they are biologically active in this form and lose activity upon irradiation and photoisomerization to their cis-isomer. Recently, cyclic azobenzenes, so-called diazocines, have emerged, which are thermodynamically more stable in their bent cis-form. Incorporation of these switches into a variety of photopharmaceuticals could convert dark-active ligands into dark-inactive ligands, which is preferred in most biological applications. This "pharmacological sign-inversion" is demonstrated for a photochromic blocker of voltage-gated potassium channels, termed CAL, and a photochromic opener of G protein-coupled inwardly rectifying potassium (GIRK) channels, termed CLOGO.


Asunto(s)
Compuestos Azo/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Luz , Bloqueadores de los Canales de Potasio/química , Potenciales de Acción/efectos de los fármacos , Compuestos Azo/farmacología , Ciclización , Diseño de Fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Células HEK293 , Humanos , Isomerismo , Lidocaína/química , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Termodinámica
5.
Glia ; 66(12): 2604-2616, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30370555

RESUMEN

Astrocytes form the largest class of glial cells in the central nervous system. They serve plenty of diverse functions that range from supporting the formation and proper operation of synapses to controlling the blood-brain barrier. For many of them, the expression of ionotropic glutamate receptors of the AMPA subtype (AMPARs) in astrocytes is of key importance. AMPARs form as macromolecular protein complexes, whose composition of the pore-lining GluA subunits and of an extensive set of core and peripheral complex constituents defines both their trafficking and gating behavior. Although astrocytic AMPARs have been reported to exhibit heterogeneous properties, their molecular composition is largely unknown. In this study, we sought to quantify the astrocytic AMPAR transcriptome during brain development and with respect to selected brain regions. Whereas the early postnatal pattern of AMPAR mRNA expression showed minor variation over time, it did show significant heterogeneity in different brain regions. Cerebellar astrocytes express a combination of AMPAR complex constituents that is remarkably distinct from the one in neocortical or hippocampal astrocytes. Our study provides a workflow and a first reference for future investigations into the molecular and functional diversity of glial AMPARs.


Asunto(s)
Astrocitos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Receptores AMPA/genética , Receptores AMPA/metabolismo , Transcriptoma/fisiología , Animales , Animales Recién Nacidos , Antígenos/genética , Antígenos/metabolismo , Astrocitos/ultraestructura , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Técnicas de Placa-Clamp , Proteoglicanos/genética , Proteoglicanos/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Xenopus laevis
7.
Mol Cell Neurosci ; 68: 331-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26363416

RESUMEN

Hepatic encephalopathy (HE) is the most common neuropsychiatric complication of acute or chronic liver failure. Clinical symptoms include cognitive and intellectual dysfunction as well as impaired motor activity and coordination. There is general consensus that increased levels of ammonia play a central role in the pathogenesis of HE. However, it is still elusive how cognitive performance including the ability to learn and memorize information is affected by ammonia at molecular levels. In the present study, we have employed a neuroglial co-culture model, which preserves neuroglial interplay but allows for cell-type specific molecular and functional analyses, to investigate glutamatergic neurotransmission under conditions of high ammonia. Chronic exposure to ammonia significantly reduced neuronal mRNA and protein expression of AMPA-subtype glutamate receptors (AMPARs), which mediate most fast excitatory neurotransmission in the brain. Surprisingly, neurons were able to fully maintain basal glutamatergic neurotransmission as recorded by AMPAR-mediated miniature excitatory postsynaptic currents (mEPSCs) even when >50% of total AMPARs were lost. However, long-lasting, activity-dependent changes in the efficacy of synaptic communication, which model the capability of the brain to learn and store information, were severely constrained. Whereas synaptic efficacy could still be depressed, an increase in synaptic strength was abolished. We conclude that neurons retain basal glutamatergic transmission at the expense of the extrasynaptic population of AMPARs, which is revealed when the extrasynaptic reserve pool is recruited in vain for synaptic potentiation. Our findings thus offer a molecular model, which might not only explain impaired synaptic plasticity in HE but also in other neurological diseases accompanied by a decrease in extrasynaptic AMPAR expression.


Asunto(s)
Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Receptores AMPA/metabolismo , Transmisión Sináptica/fisiología , Amoníaco/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Canales de Calcio/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/citología , L-Lactato Deshidrogenasa/metabolismo , N-Metilaspartato/farmacología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Ratas , Receptores AMPA/genética , Transmisión Sináptica/efectos de los fármacos
8.
Mol Cell Neurosci ; 56: 10-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23403072

RESUMEN

Besides mediating most of the fast excitatory neurotransmission in the mammalian CNS, ionotropic glutamate receptors of the AMPA subtype (AMPARs) serve highly diverse functions in brain development controlling neuronal migration, synaptic growth, and synaptic maturation. Pioneering proteomic studies suggest that this functional diversity is met by a great molecular complexity in native AMPAR composition. Here, we have investigated the expression patterns of two recently identified AMPAR constituents, the cornichon homologues CNIH-2 and CNIH-3, and their assembly with the AMPAR core subunits GluA1-4 in developing rat brain. Unlike GluA1-4 expression, which is up-regulated during postnatal brain development, the two cornichon homologues show maximum mRNA and protein expression early after birth, which then decline towards adulthood. Despite rather reciprocal expression profiles, the overall ratio of CNIH-2/3 complexed with GluAs remains constant throughout development. Our data reveal an excess amount of AMPAR-free CNIH-2/3 early in development, which might serve the evolutionarily conserved role of cornichon as a cargo exporter. With progressing development, however, the amount of AMPAR-free CNIH-2/3 subsides, whereas the one being integrated into AMPAR complexes increases. Hence, the cornichon homologues CNIH-2/3 gain importance in their role as auxiliary subunits of native AMPARs during ontogeny, which reflects their functional evolution in phylogeny.


Asunto(s)
Receptores AMPA/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Filogenia , Unión Proteica , Multimerización de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/genética
9.
Br J Pharmacol ; 181(12): 1734-1756, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38157867

RESUMEN

BACKGROUND AND PURPOSE: Neuropathic pain affects up to 10% of the global population and is caused by an injury or a disease affecting the somatosensory, peripheral, or central nervous system. NP is characterized by chronic, severe and opioid-resistant properties. Therefore, its clinical management remains very challenging. The N-type voltage-gated calcium channel, Cav2.2, is a validated target for therapeutic intervention in chronic and neuropathic pain. The conotoxin ziconotide (Prialt®) is an FDA-approved drug that blocks Cav2.2 channel but needs to be administered intrathecally. Thus, although being principally efficient, the required application route is very much in disfavour. EXPERIMENTAL APPROACH AND KEY RESULTS: Here, we describe an orally available drug candidate, RD2, which competes with ziconotide binding to Cav2.2 at nanomolar concentrations and inhibits Cav2.2 almost completely reversible. Other voltage-gated calcium channel subtypes, like Cav1.2 and Cav3.2, were affected by RD2 only at concentrations higher than 10 µM. Data from sciatic inflammatory neuritis rat model demonstrated the in vivo proof of concept, as low-dose RD2 (5 mg·kg-1) administered orally alleviated neuropathic pain compared with vehicle controls. High-dose RD2 (50 mg·kg-1) was necessary to reduce pain sensation in acute thermal response assessed by the tail flick test. CONCLUSIONS AND IMPLICATIONS: Taken together, these results demonstrate that RD2 has antiallodynic properties. RD2 is orally available, which is the most convenient application form for patients and caregivers. The surprising and novel result from standard receptor screens opens the room for further optimization into new promising drug candidates, which address an unmet medical need.


Asunto(s)
Bloqueadores de los Canales de Calcio , Canales de Calcio Tipo N , Neuralgia , Animales , Humanos , Masculino , Ratones , Ratas , Administración Oral , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo N/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ratones Endogámicos C57BL , Neuralgia/tratamiento farmacológico , omega-Conotoxinas/administración & dosificación , omega-Conotoxinas/farmacología , omega-Conotoxinas/uso terapéutico , Ratas Endogámicas Lew
10.
Arch Biochem Biophys ; 536(2): 183-8, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23624147

RESUMEN

Hepatic encephalopathy (HE)(1) is a common neuropsychiatric complication of both acute and chronic liver disease. Clinical symptoms may include motor disturbances and cognitive dysfunction. Available animal models of HE mimic the deficits in cognitive performance including the impaired ability to learn and memorize information. This review explores the question how HE might affect cognitive functions at molecular levels. Both acute and chronic models of HE constrain the plasticity of glutamatergic neurotransmission. Thus, long-lasting activity-dependent changes in synaptic efficiency, known as long-term potentiation (LTP) and long-term depression (LTD) are significantly impeded. We discuss molecules and signal transduction pathways of LTP and LTD that are targeted by experimental HE, with a focus on ionotropic glutamate receptors of the AMPA-subtype. Finally, a novel strategy of functional proteomic analysis is presented, which, if applied differentially, may provide molecular insight into disease-related dysfunction of membrane protein complexes, i.e. disturbed ionotropic glutamate receptor signaling in HE.


Asunto(s)
Encéfalo/fisiopatología , Encefalopatía Hepática/fisiopatología , Plasticidad Neuronal , Sinapsis/patología , Amoníaco/metabolismo , Animales , Encéfalo/metabolismo , Encefalopatía Hepática/metabolismo , Humanos , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
11.
Physiol Rep ; 11(11): e15670, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37291479

RESUMEN

Ventricular repolarization shows notable sex-specificity, with female sex being associated with longer QT-intervals in electrocardiography irrespective of the species studied. From a clinical point of view, women are at a greater risk for drug-induced torsade de pointes and symptomatic long-QT syndrome. Here, we present an optical mapping (OM) approach to reveal sex-specific action potential (AP) heterogeneity in a slice preparation of mouse hearts. Left ventricular epicardial repolarization in female versus male mice shows longer and, interindividually, more variable AP duration (APD), yielding a less prominent transmural APD gradient. By combining OM with mathematical modeling, we suggest a significant role of IKto,f and IKur in AP broadening in females. Other transmembrane currents, including INaL , only marginally affect basal APD. As in many cardiac pathophysiologies, increasing [Ca2+ ]i poses a risk for arrhythmia, the response of AP morphology to enhanced activation of L-type calcium channels (LTCC) was assessed in a sex-selective manner. Both APD and its variation increased significantly more in female versus male mice after pharmacological LTCC activation, which we hypothesize to be due to sex-specific INaL expression based on mathematical modeling. Altogether, we demonstrate a more delayed repolarization of LV epicardium, a leveled LV transmural APD gradient, and a more pronounced epicardial APD response to Ca2+ influx in females versus males. Mathematical modeling quantifies the relative contributions of selected ionic currents to sex-specific AP morphology under normal and pathophysiological conditions.


Asunto(s)
Electrocardiografía , Ventrículos Cardíacos , Femenino , Masculino , Animales , Ratones , Ventrículos Cardíacos/metabolismo , Arritmias Cardíacas/metabolismo , Pericardio , Potenciales de Acción
12.
Front Cardiovasc Med ; 10: 1171831, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37252113

RESUMEN

Introduction: Platelet activation and thrombus formation is crucial for hemostasis, but also trigger arterial thrombosis. Calcium mobilization plays an important role in platelet activation, because many cellular processes depend on the level of intracellular Ca2+ ([Ca2+](i)), such as integrin activation, degranulation, cytoskeletal reorganization. Different modulators of Ca2+ signaling have been implied, such as STIM1, Orai1, CyPA, SGK1, etc. Also, the N-methyl-D-aspartate receptor (NMDAR) was identified to contribute to Ca2+ signaling in platelets. However, the role of the NMDAR in thrombus formation is not well defined. Methods: In vitro and in vivo analysis of platelet-specific NMDAR knock-out mice. Results: In this study, we analyzed Grin1fl/fl-Pf4-Cre+ mice with a platelet-specific knock-out of the essential GluN1 subunit of the NMDAR. We found reduced store-operated Ca2+ entry (SOCE), but unaltered store release in GluN1-deficient platelets. Defective SOCE resulted in reduced Src and PKC substrate phosphorylation following stimulation of glycoprotein (GP)VI or the thrombin receptor PAR4 followed by decreased integrin activation but unaltered degranulation. Consequently, thrombus formation on collagen under flow conditions was reduced ex vivo, and Grin1fl/fl-Pf4-Cre+ mice were protected against arterial thrombosis. Results from human platelets treated with the NMDAR antagonist MK-801 revealed a crucial role of the NMDAR in integrin activation and Ca2+ homeostasis in human platelets as well. Conclusion: NMDAR signaling is important for SOCE in platelets and contributes to platelet activation and arterial thrombosis. Thus, the NMDAR represents a novel target for anti-platelet therapy in cardiovascular disease (CVD).

13.
Sci Rep ; 13(1): 5693, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029160

RESUMEN

Malignant ventricular arrhythmias (VA) after acute myocardial infarction remain a major threat. Aim of this study was to characterize the electrophysiological and autonomic sequelae of cardiac ischemia and reperfusion (I/R) in mice during the first week post incident. Left ventricular function was serially assessed using transthoracic echocardiography. VA were quantified by telemetric electrocardiogram (ECG) recordings and electrophysiological studies on the 2nd and 7th day after I/R. Cardiac autonomic function was evaluated by heart rate variability (HRV) and heart rate turbulence (HRT). Infarct size was quantified by planimetric measures. I/R caused significant myocardial scarring and diminished left ventricular ejection fraction. The ECG intervals QRS, QT, QTc, and JTc were prolonged in I/R mice. Both spontaneous VA scored higher and the inducibility of VA was raised in I/R mice. An analysis of HRV and HRT indicated a relative reduction in parasympathetic activity and disturbed baroreflex sensitivity up to 7 days after I/R. In summary, during the first week after I/R, the murine heart reflects essential features of the human heart after myocardial infarction, including a greater vulnerability for VA and a decreased parasympathetic tone accompanied by decelerated depolarization and repolarization parameters.


Asunto(s)
Enfermedad de la Arteria Coronaria , Infarto del Miocardio , Isquemia Miocárdica , Humanos , Animales , Ratones , Volumen Sistólico , Función Ventricular Izquierda , Isquemia Miocárdica/complicaciones , Electrocardiografía , Enfermedad de la Arteria Coronaria/complicaciones , Arritmias Cardíacas/complicaciones , Reperfusión Miocárdica , Frecuencia Cardíaca/fisiología
14.
Acta Physiol (Oxf) ; 239(2): e14004, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37227741

RESUMEN

AIM: Cardiac pathologies are accompanied by alterations in substrate metabolism, and extracellular flux analysis is a standard tool to investigate metabolic disturbances, especially in immortalized cell lines. However, preparations of primary cells, such as adult cardiomyocytes require enzymatic dissociation and cultivation affecting metabolism. Therefore, we developed a flux analyzer-based method for the assessment of substrate metabolism in intact vibratome-sliced mouse heart tissue. METHODS: Oxygen consumption rates were determined using a Seahorse XFe24-analyzer and "islet capture plates." We demonstrate that tissue slices are suitable for extracellular flux analysis and metabolize both free fatty acids (FFA) and glucose/glutamine. Functional integrity of tissue slices was proven by optical mapping-based assessment of action potentials. In a proof-of-principle approach, the sensitivity of the method was tested by analyzing substrate metabolism in the remote myocardium after myocardial infarction (I/R). RESULTS: Here, I/R increased uncoupled OCR compared with sham animals indicating a stimulated metabolic capacity. This increase was caused by a higher glucose/glutamine metabolism, whereas FFA oxidation was unchanged. CONCLUSION: In conclusion, we describe a novel method to analyze cardiac substrate metabolism in intact cardiac tissue slices by extracellular flux analysis. The proof-of-principle experiment demonstrated that this approach has a sensitivity allowing the investigation of pathophysiologically relevant disturbances in cardiac substrate metabolism.


Asunto(s)
Glutamina , Miocardio , Animales , Ratones , Glutamina/metabolismo , Miocardio/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Miocitos Cardíacos/metabolismo , Consumo de Oxígeno/fisiología
15.
Science ; 381(6660): 897-906, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37616346

RESUMEN

Aging is a major risk factor for impaired cardiovascular health. Because the aging myocardium is characterized by microcirculatory dysfunction, and because nerves align with vessels, we assessed the impact of aging on the cardiac neurovascular interface. We report that aging reduces nerve density in the ventricle and dysregulates vascular-derived neuroregulatory genes. Aging down-regulates microRNA 145 (miR-145) and derepresses the neurorepulsive factor semaphorin-3A. miR-145 deletion, which increased Sema3a expression or endothelial Sema3a overexpression, reduced axon density, mimicking the aged-heart phenotype. Removal of senescent cells, which accumulated with chronological age in parallel to the decline in nerve density, rescued age-induced denervation, reversed Sema3a expression, preserved heart rate patterns, and reduced electrical instability. These data suggest that senescence-mediated regulation of nerve density contributes to age-associated cardiac dysfunction.


Asunto(s)
Envejecimiento , Senescencia Celular , Corazón , MicroARNs , Densidad Microvascular , Miocardio , Semaforina-3A , Corazón/inervación , Microcirculación , MicroARNs/genética , MicroARNs/metabolismo , Semaforina-3A/genética , Animales , Ratones , Envejecimiento/genética , Envejecimiento/patología , Masculino , Ratones Endogámicos C57BL , Senescencia Celular/genética , Miocardio/patología , Axones
16.
Cell Physiol Biochem ; 30(5): 1227-40, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23075627

RESUMEN

BACKGROUND/AIMS: The D553N mutation located in the C-linker of the cardiac pacemaker channel HCN4 is thought to cause sino-atrial dysfunction via a pronounced dominant-negative trafficking defect. Since HCN4 mutations usually have a minor defect in channel gating, it was our aim to further characterize the disease causing mechanism of D553N. METHODS: Fluorescence microscopy, FACS, TEVC and patch-clamp recordings were performed to characterize D553N. RESULTS: Surprisingly, we found that D553N channels reach the plasma membrane and have no apparent trafficking defect. Co-expression of D553N with HCN4 also revealed no dominant-negative effect on wild-type channels. Consistent with the normal cell surface expression of D553N, it was possible to extensively characterize D553N mutants in Xenopus oocytes and mammalian cells. D553N channels generate currents with reduced amplitude, while the kinetics of activation and deactivation are not altered. While the regulation of D553N by tyrosine kinases is normal, we observed a change in the cAMP regulation which however cannot account for the strong loss-of-function of the mutant. CONCLUSION: The pronounced current reduction and the regular surface expression indicate a major gating defect of the C-linker gate. We hypothesize that the D553N mutation stabilizes a previously reported salt bridge important for the gating of the channel.


Asunto(s)
Bradicardia/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Activación del Canal Iónico , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Canales de Potasio/genética , Canales de Potasio/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Proteínas Musculares/química , Canales de Potasio/química , Xenopus
17.
Auton Neurosci ; 241: 103009, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35753247

RESUMEN

BACKGROUND: The importance of peripheral chemoreceptors for cardiorespiratory neural control is known for decades. Pure oxygen inhalation deactivates chemoreceptors and increases parasympathetic outflow. However, the relationship between autonomic nervous system (ANS) activation and resulting respiratory as well as heart rate (HR) dynamics is still not fully understood. METHODS: In young adults the impact of (1) 100 % pure oxygen inhalation (hyperoxic cardiac chemoreflex sensitivity (CHRS) testing), (2) the cold face test (CFT) and (3) the cold pressor test (CPT) on heart rate variability (HRV), hemodynamics and respiratory rate was investigated in randomized order. Baseline ANS outflow was determined assessing respiratory sinus arrhythmia via deep breathing, baroreflex sensitivity and HRV. RESULTS: Baseline ANS outflow was normal in all participants (23 ± 1 years, 7 females, 3 males). Hyperoxic CHRS testing decreased HR (after 60 ± 3 vs before 63 ± 3 min-1, p = 0.004), while increasing total peripheral resistance (1053 ± 87 vs 988 ± 76 dyne*s + m2/cm5, p = 0.02) and mean arterial blood pressure (93 ± 4 vs 91 ± 4 mm Hg, p = 0.02). HRV indicated increased parasympathetic outflow after hyperoxic CHRS testing accompanied by a decrease in respiratory rate (15 ± 1vs 19 ± 1 min-1, p = 0.001). In contrast, neither CFT nor CPT altered the respiratory rate (18 ± 1 vs 18 ± 2 min-1, p = 0.38 and 18 ± 1 vs 18 ± 1 min-1, p = 0.84, respectively). CONCLUSION: Changes in HR characteristics during deactivation of peripheral chemoreceptors but not during the CFT and CPT are related with a decrease in respiratory rate. This highlights the need of respiratory rate assessment when evaluating adaptations of cardiorespiratory chemoreceptor control.


Asunto(s)
Frecuencia Respiratoria , Sistema Nervioso Simpático , Presión Sanguínea/fisiología , Células Quimiorreceptoras/fisiología , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Oxígeno , Adulto Joven
18.
Cell Chem Biol ; 28(10): 1474-1488.e7, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34118188

RESUMEN

Dextromethorphan (DXM) acts as cough suppressant via its central action. Cell-protective effects of this drug have been reported in peripheral tissues, making DXM potentially useful for treatment of several common human diseases, such as type 2 diabetes mellitus (T2DM). Pancreatic islets are among the peripheral tissues that positively respond to DXM, and anti-diabetic effects of DXM were observed in two placebo-controlled, randomized clinical trials in humans with T2DM. Since these effects were associated with central side effects, we here developed chemical derivatives of DXM that pass the blood-brain barrier to a significantly lower extent than the original drug. We show that basic nitrogen-containing residues block central adverse events of DXM without reducing its anti-diabetic effects, including the protection of human pancreatic islets from cell death. These results show how to chemically modify DXM, and possibly other morphinans, as to exclude central side effects, while targeting peripheral tissues, such as pancreatic islets.


Asunto(s)
Glucemia/análisis , Dextrometorfano/farmacología , Hipoglucemiantes/farmacología , Islotes Pancreáticos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Dextrometorfano/análogos & derivados , Dextrometorfano/metabolismo , Dextrometorfano/uso terapéutico , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Diseño de Fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones Endogámicos C57BL
19.
Neuron ; 52(6): 1027-36, 2006 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17178405

RESUMEN

Hyperpolarization-activated, cyclic-nucleotide-gated (HCN) channels mediate the depolarizing cation current (termed I(h) or I(f)) that initiates spontaneous rhythmic activity in heart and brain. This function critically depends on the reliable opening of HCN channels in the subthreshold voltage-range. Here we show that activation of HCN channels at physiologically relevant voltages requires interaction with phosphoinositides such as phosphatidylinositol-4,5-bisphosphate (PIP(2)). PIP(2) acts as a ligand that allosterically opens HCN channels by shifting voltage-dependent channel activation approximately 20 mV toward depolarized potentials. Allosteric gating by PIP(2) occurs in all HCN subtypes and is independent of the action of cyclic nucleotides. In CNS neurons and cardiomyocytes, enzymatic degradation of phospholipids results in reduced channel activation and slowing of the spontaneous firing rate. These results demonstrate that gating by phospholipids is essential for the pacemaking activity of HCN channels in cardiac and neuronal rhythmogenesis.


Asunto(s)
Relojes Biológicos/fisiología , Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Neuronas/fisiología , Fosfatidilinositoles/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Androstadienos/farmacología , Animales , Relojes Biológicos/efectos de los fármacos , Encéfalo/citología , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Embrión de Mamíferos , Embrión no Mamífero , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Mutación/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/efectos de la radiación , Neuronas/efectos de los fármacos , Oocitos , Técnicas de Placa-Clamp/métodos , Fosfatidilinositol 4,5-Difosfato/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Canales de Potasio , Pirimidinas/farmacología , Wortmanina , Xenopus
20.
Neuron ; 49(5): 697-706, 2006 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-16504945

RESUMEN

The voltage-gated potassium (Kv) channel subunit Kv1.1 is a major constituent of presynaptic A-type channels that modulate synaptic transmission in CNS neurons. Here, we show that Kv1.1-containing channels are complexed with Lgi1, the functionally unassigned product of the leucine-rich glioma inactivated gene 1 (LGI1), which is causative for an autosomal dominant form of lateral temporal lobe epilepsy (ADLTE). In the hippocampal formation, both Kv1.1 and Lgi1 are coassembled with Kv1.4 and Kvbeta1 in axonal terminals. In A-type channels composed of these subunits, Lgi1 selectively prevents N-type inactivation mediated by the Kvbeta1 subunit. In contrast, defective Lgi1 molecules identified in ADLTE patients fail to exert this effect resulting in channels with rapid inactivation kinetics. The results establish Lgi1 as a novel subunit of Kv1.1-associated protein complexes and suggest that changes in inactivation gating of presynaptic A-type channels may promote epileptic activity.


Asunto(s)
Encéfalo/metabolismo , Canal de Potasio Kv.1.1/fisiología , Canal de Potasio Kv.1.2/fisiología , Inhibición Neural/fisiología , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting/métodos , Encéfalo/citología , Química Encefálica , Membrana Celular/metabolismo , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Humanos , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intracelular , Espectrometría de Masas/métodos , Potenciales de la Membrana/fisiología , Mutagénesis/fisiología , Mutación , Oocitos , Técnicas de Placa-Clamp/métodos , Conformación Proteica , Ratas , Alineación de Secuencia , Tinción con Nitrato de Plata/métodos , Transfección/métodos , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA