Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34021086

RESUMEN

In cardiomyocytes, NaV1.5 channels mediate initiation and fast propagation of action potentials. The Ca2+-binding protein calmodulin (CaM) serves as a de facto subunit of NaV1.5. Genetic studies and atomic structures suggest that this interaction is pathophysiologically critical, as human mutations within the NaV1.5 carboxy-terminus that disrupt CaM binding are linked to distinct forms of life-threatening arrhythmias, including long QT syndrome 3, a "gain-of-function" defect, and Brugada syndrome, a "loss-of-function" phenotype. Yet, how a common disruption in CaM binding engenders divergent effects on NaV1.5 gating is not fully understood, though vital for elucidating arrhythmogenic mechanisms and for developing new therapies. Here, using extensive single-channel analysis, we find that the disruption of Ca2+-free CaM preassociation with NaV1.5 exerts two disparate effects: 1) a decrease in the peak open probability and 2) an increase in persistent NaV openings. Mechanistically, these effects arise from a CaM-dependent switch in the NaV inactivation mechanism. Specifically, CaM-bound channels preferentially inactivate from the open state, while those devoid of CaM exhibit enhanced closed-state inactivation. Further enriching this scheme, for certain mutant NaV1.5, local Ca2+ fluctuations elicit a rapid recruitment of CaM that reverses the increase in persistent Na current, a factor that may promote beat-to-beat variability in late Na current. In all, these findings identify the elementary mechanism of CaM regulation of NaV1.5 and, in so doing, unravel a noncanonical role for CaM in tuning ion channel gating. Furthermore, our results furnish an in-depth molecular framework for understanding complex arrhythmogenic phenotypes of NaV1.5 channelopathies.


Asunto(s)
Potenciales de Acción/genética , Calcio/metabolismo , Calmodulina/química , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/química , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Sitios de Unión , Señalización del Calcio , Calmodulina/genética , Calmodulina/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Células HEK293 , Humanos , Activación del Canal Iónico , Cinética , Modelos Moleculares , Mutación , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sodio/metabolismo
2.
J Biol Chem ; 296: 100502, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33667546

RESUMEN

Ca2+/calmodulin-dependent inactivation (CDI) of CaV channels is a critical regulatory process that tunes the kinetics of Ca2+ entry for different cell types and physiologic responses. CDI is mediated by calmodulin (CaM), which is bound to the IQ domain of the CaV carboxy tail. This modulatory process is tailored by alternative splicing such that select splice variants of CaV1.3 and CaV1.4 contain a long distal carboxy tail (DCT). The DCT harbors an inhibitor of CDI (ICDI) module that competitively displaces CaM from the IQ domain, thereby diminishing CDI. While this overall mechanism is now well described, the detailed interactions required for ICDI binding to the IQ domain are yet to be elucidated. Here, we perform alanine-scanning mutagenesis of the IQ and ICDI domains and evaluate the contribution of neighboring regions to CDI inhibition. Through FRET binding analysis, we identify functionally relevant residues within the CaV1.3 IQ domain and the CaV1.4 ICDI and nearby A region, which are required for high-affinity IQ/ICDI binding. Importantly, patch-clamp recordings demonstrate that disruption of this interaction commensurately diminishes ICDI function resulting in the re-emergence of CDI in mutant channels. Furthermore, CaV1.2 channels harbor a homologous DCT; however, the ICDI region of this channel does not appear to appreciably modulate CaV1.2 CDI. Yet coexpression of CaV1.2 ICDI with select CaV1.3 splice variants significantly disrupts CDI, implicating a cross-channel modulatory scheme in cells expressing both channel subtypes. In all, these findings provide new insights into a molecular rheostat that fine-tunes Ca2+-entry and supports normal neuronal and cardiac function.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Caveolina 1/metabolismo , Activación del Canal Iónico , Mutación , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Células Cultivadas , Humanos , Cinética , Unión Proteica , Relación Estructura-Actividad
3.
Paediatr Child Health ; 24(2): e74-e77, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30996610

RESUMEN

BACKGROUND: Referrals to paediatric endocrine clinics for short stature are common. Height velocity (HV) is an essential component of the evaluation of short stature as growth deceleration often reflects an underlying paediatric endocrine diagnosis (PED). Access to previous measurements facilitates prompt calculation of HV. OBJECTIVE: To determine the availability of previous measurements at time of referral for short stature and to determine predictors of a PED. METHODS: A retrospective chart review was performed on all referrals for short stature to a single paediatric endocrinologist between January 2008 and December 2014. Standard practice following receipt of a referral for short stature included repeated requests to the referring physician for previous measurements. RESULTS: A total of 324 charts of patients aged 11 months to 18 years were reviewed and 286 were eligible for inclusion. Previous measurements were available in 72.4%, and 44.8% of these were found to have a PED. There was a significant relation between HV<25th percentile and a PED (P<0.0001) and between height deficit (HD) and a PED (P<0.0001). Logistic regression analysis showed that a HV<25th percentile and a HD>2 standard deviations, increased the odds of PED by a factor of 5.12 (P<0.001) and 1.39 (P<0.005), respectively. CONCLUSION: HV is a significant predictor of a PED. Our higher rate of previous measurement availability is likely due to our effective referral screening protocol. The availability of these measurements, which are essential for HV calculation, are likely to reduce delays in diagnosis and management.

4.
J Gen Physiol ; 150(12): 1688-1701, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30470716

RESUMEN

L-type calcium channels (LTCCs) are critical elements of normal cardiac function, playing a major role in orchestrating cardiac electrical activity and initiating downstream signaling processes. LTCCs thus use feedback mechanisms to precisely control calcium (Ca2+) entry into cells. Of these, Ca2+-dependent inactivation (CDI) is significant because it shapes cardiac action potential duration and is essential for normal cardiac rhythm. This important form of regulation is mediated by a resident Ca2+ sensor, calmodulin (CaM), which is comprised of two lobes that are each capable of responding to spatially distinct Ca2+ sources. Disruption of CaM-mediated CDI leads to severe forms of long-QT syndrome (LQTS) and life-threatening arrhythmias. Thus, a model capable of capturing the nuances of CaM-mediated CDI would facilitate increased understanding of cardiac (patho)physiology. However, one critical barrier to achieving a detailed kinetic model of CDI has been the lack of quantitative data characterizing CDI as a function of Ca2+ This data deficit stems from the experimental challenge of uncoupling the effect of channel gating on Ca2+ entry. To overcome this obstacle, we use photo-uncaging of Ca2+ to deliver a measurable Ca2+ input to CaM/LTCCs, while simultaneously recording CDI. Moreover, we use engineered CaMs with Ca2+ binding restricted to a single lobe, to isolate the kinetic response of each lobe. These high-resolution measurements enable us to build mathematical models for each lobe of CaM, which we use as building blocks for a full-scale bilobal model of CDI. Finally, we use this model to probe the pathogenesis of LQTS associated with mutations in CaM (calmodulinopathies). Each of these models accurately recapitulates the kinetics and steady-state properties of CDI in both physiological and pathological states, thus offering powerful new insights into the mechanistic alterations underlying cardiac arrhythmias.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Calcio/metabolismo , Calmodulina/metabolismo , Modelos Químicos , Calmodulina/genética , Humanos , Síndrome de QT Prolongado/genética
5.
Elife ; 72018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30198845

RESUMEN

Calmodulin (CaM) serves as a pervasive regulatory subunit of CaV1, CaV2, and NaV1 channels, exploiting a functionally conserved carboxy-tail element to afford dynamic Ca2+-feedback of cellular excitability in neurons and cardiomyocytes. Yet this modularity counters functional adaptability, as global changes in ambient CaM indiscriminately alter its targets. Here, we demonstrate that two structurally unrelated proteins, SH3 and cysteine-rich domain (stac) and fibroblast growth factor homologous factors (fhf) selectively diminish Ca2+/CaM-regulation of CaV1 and NaV1 families, respectively. The two proteins operate on allosteric sites within upstream portions of respective channel carboxy-tails, distinct from the CaM-binding interface. Generalizing this mechanism, insertion of a short RxxK binding motif into CaV1.3 carboxy-tail confers synthetic switching of CaM regulation by Mona SH3 domain. Overall, our findings identify a general class of auxiliary proteins that modify Ca2+/CaM signaling to individual targets allowing spatial and temporal orchestration of feedback, and outline strategies for engineering Ca2+/CaM signaling to individual targets.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Retroalimentación Fisiológica , Canales de Sodio/metabolismo , Potenciales de Acción , Regulación Alostérica , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Canales de Calcio/química , Calmodulina/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Mutagénesis , Proteínas del Tejido Nervioso , Unión Proteica , Dominios Proteicos , Ingeniería de Proteínas , Ratas , Transducción de Señal
6.
J Gen Physiol ; 150(8): 1145-1161, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29950399

RESUMEN

CaV1.1 is essential for skeletal muscle excitation-contraction coupling. Its functional expression is tuned by numerous regulatory proteins, yet underlying modulatory mechanisms remain ambiguous as CaV1.1 fails to function in heterologous systems. In this study, by dissecting channel trafficking versus gating, we evaluated the requirements for functional CaV1.1 in heterologous systems. Although coexpression of the auxiliary ß subunit is sufficient for surface-membrane localization, this baseline trafficking is weak, and channels elicit a diminished open probability. The regulatory proteins calmodulin and stac3 independently enhance channel trafficking and gating via their interaction with the CaV1.1 carboxy terminus. Myopathic stac3 mutations weaken channel binding and diminish trafficking. Our findings demonstrate that multiple regulatory proteins orchestrate CaV1.1 function via duplex mechanisms. Our work also furnishes insights into the pathophysiology of stac3-associated congenital myopathy and reveals novel avenues for pharmacological intervention.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calmodulina/metabolismo , Células HEK293 , Humanos , Enfermedades Musculares/congénito , Enfermedades Musculares/metabolismo , Técnicas de Placa-Clamp
7.
J Asthma Allergy ; 11: 111-120, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29950871

RESUMEN

Anaphylaxis is a severe and potentially life-threatening allergic reaction. There are numerous potential causes, with food allergy being the leading cause in children and the focus of this review. Most reactions involve an IgE-mediated mechanism, although non-IgE-mediated and nonimmunologic reactions can occur. Various cofactors to be discussed can place certain individuals at an increased risk of severe or fatal anaphylaxis. The clinical manifestations of anaphylaxis are broad and may involve multiple body systems. Diagnosis of food-related anaphylaxis is primarily based on signs and symptoms and supported, wherever possible, by identification and confirmation of a culprit food allergen. First-line treatment of anaphylaxis is intramuscular administration of epinephrine. Long-term management is generally focused on strict allergen avoidance and more recently on food desensitization using immunotherapy. This review provides an overview of anaphylaxis with a specific focus on food allergy.

8.
Biophys J ; 114(9): 2231-2242, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742416

RESUMEN

Cell volume regulation is fundamentally important in phenomena such as cell growth, proliferation, tissue homeostasis, and embryogenesis. How the cell size is set, maintained, and changed over a cell's lifetime is not well understood. In this work we focus on how the volume of nonexcitable tissue cells is coupled to the cell membrane electrical potential and the concentrations of membrane-permeable ions in the cell environment. Specifically, we demonstrate that a sudden cell depolarization using the whole-cell patch clamp results in a 50% increase in cell volume, whereas hyperpolarization results in a slight volume decrease. We find that cell volume can be partially controlled by changing the chloride or the sodium/potassium concentrations in the extracellular environment while maintaining a constant external osmotic pressure. Depletion of external chloride leads to a volume decrease in suspended HN31 cells. Introducing cells to a high-potassium solution causes volume increase up to 50%. Cell volume is also influenced by cortical tension: actin depolymerization leads to cell volume increase. We present an electrophysiology model of water dynamics driven by changes in membrane potential and the concentrations of permeable ions in the cells surrounding. The model quantitatively predicts that the cell volume is directly proportional to the intracellular protein content.


Asunto(s)
Tamaño de la Célula , Fenómenos Electrofisiológicos , Actinas/química , Línea Celular Tumoral , Cloruros/metabolismo , Espacio Extracelular/metabolismo , Humanos , Espacio Intracelular/metabolismo , Potasio/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Sodio/metabolismo
9.
J Mol Cell Cardiol ; 119: 64-74, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29684406

RESUMEN

Dysregulation of L-type Ca2+ channels (LTCCs) underlies numerous cardiac pathologies. Understanding their modulation with high fidelity relies on investigating LTCCs in their native environment with intact interacting proteins. Such studies benefit from genetic manipulation of endogenous channels in cardiomyocytes, which often proves cumbersome in mammalian models. Drosophila melanogaster, however, offers a potentially efficient alternative as it possesses a relatively simple heart, is genetically pliable, and expresses well-conserved genes. Fluorescence in situ hybridization confirmed an abundance of Ca-α1D and Ca-α1T mRNA in fly myocardium, which encode subunits that specify hetero-oligomeric channels homologous to mammalian LTCCs and T-type Ca2+ channels, respectively. Cardiac-specific knockdown of Ca-α1D via interfering RNA abolished cardiac contraction, suggesting Ca-α1D (i.e. A1D) represents the primary functioning Ca2+ channel in Drosophila hearts. Moreover, we successfully isolated viable single cardiomyocytes and recorded Ca2+ currents via patch clamping, a feat never before accomplished with the fly model. The profile of Ca2+ currents recorded in individual cells when Ca2+ channels were hypomorphic, absent, or under selective LTCC blockage by nifedipine, additionally confirmed the predominance of A1D current across all activation voltages. T-type current, activated at more negative voltages, was also detected. Lastly, A1D channels displayed Ca2+-dependent inactivation, a critical negative feedback mechanism of LTCCs, and the current through them was augmented by forskolin, an activator of the protein kinase A pathway. In sum, the Drosophila heart possesses a conserved compendium of Ca2+ channels, suggesting that the fly may serve as a robust and effective platform for studying cardiac channelopathies.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canalopatías/metabolismo , Drosophila melanogaster/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Cardiotónicos/farmacología , Colforsina/farmacología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Retroalimentación Fisiológica/fisiología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Hibridación Fluorescente in Situ , Masculino , Contracción Miocárdica/fisiología , Nifedipino/farmacología , Técnicas de Placa-Clamp
10.
Proc Natl Acad Sci U S A ; 115(13): E3026-E3035, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29531055

RESUMEN

Calmodulin (CaM) regulation of voltage-gated calcium (CaV) channels is a powerful Ca2+ feedback mechanism that adjusts Ca2+ influx, affording rich mechanistic insights into Ca2+ decoding. CaM possesses a dual-lobed architecture, a salient feature of the myriad Ca2+-sensing proteins, where two homologous lobes that recognize similar targets hint at redundant signaling mechanisms. Here, by tethering CaM lobes, we demonstrate that bilobal architecture is obligatory for signaling to CaV channels. With one lobe bound, CaV carboxy tail rearranges itself, resulting in a preinhibited configuration precluded from Ca2+ feedback. Reconstitution of two lobes, even as separate molecules, relieves preinhibition and restores Ca2+ feedback. CaV channels thus detect the coincident binding of two Ca2+-free lobes to promote channel opening, a molecular implementation of a logical NOR operation that processes spatiotemporal Ca2+ signals bifurcated by CaM lobes. Overall, a unified scheme of CaV channel regulation by CaM now emerges, and our findings highlight the versatility of CaM to perform exquisite Ca2+ computations.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Calmodulina/metabolismo , Activación del Canal Iónico/fisiología , Secuencia de Aminoácidos , Animales , Canales de Calcio/química , Calmodulina/química , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Ratas , Homología de Secuencia , Transducción de Señal
11.
Hear Res ; 344: 207-222, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28011084

RESUMEN

Functional organization is a key feature of the neocortex that often guides studies of sensory processing, development, and plasticity. Tonotopy, which arises from the transduction properties of the cochlea, is the most widely studied organizational feature in auditory cortex; however, in order to process complex sounds, cortical regions are likely specialized for higher order features. Here, motivated by the prevalence of frequency modulations in mouse ultrasonic vocalizations and aided by the use of a multiscale imaging approach, we uncover a functional organization across the extent of auditory cortex for the rate of frequency modulated (FM) sweeps. In particular, using two-photon Ca2+ imaging of layer 2/3 neurons, we identify a tone-insensitive region at the border of AI and AAF. This central sweep region behaves fundamentally differently from nearby neurons in AI and AII, responding preferentially to fast FM sweeps but not to tones or bandlimited noise. Together these findings define a second dimension of organization in the mouse auditory cortex for sweep rate complementary to that of tone frequency.


Asunto(s)
Corteza Auditiva/fisiología , Técnicas Biosensibles , Mapeo Encefálico/métodos , Microscopía de Fluorescencia por Excitación Multifotónica , Percepción de la Altura Tonal , Estimulación Acústica , Animales , Corteza Auditiva/metabolismo , Calcio/metabolismo , Potenciales Evocados Auditivos , Genes Reporteros , Ratones Transgénicos , Plasticidad Neuronal , Factores de Tiempo
12.
Circ Res ; 120(1): 39-48, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-27765793

RESUMEN

RATIONALE: Calmodulinopathies comprise a new category of potentially life-threatening genetic arrhythmia syndromes capable of producing severe long-QT syndrome (LQTS) with mutations involving CALM1, CALM2, or CALM3. The underlying basis of this form of LQTS is a disruption of Ca2+/calmodulin (CaM)-dependent inactivation of L-type Ca2+ channels. OBJECTIVE: To gain insight into the mechanistic underpinnings of calmodulinopathies and devise new therapeutic strategies for the treatment of this form of LQTS. METHODS AND RESULTS: We generated and characterized the functional properties of induced pluripotent stem cell-derived cardiomyocytes from a patient with D130G-CALM2-mediated LQTS, thus creating a platform with which to devise and test novel therapeutic strategies. The patient-derived induced pluripotent stem cell-derived cardiomyocytes display (1) significantly prolonged action potentials, (2) disrupted Ca2+ cycling properties, and (3) diminished Ca2+/CaM-dependent inactivation of L-type Ca2+ channels. Next, taking advantage of the fact that calmodulinopathy patients harbor a mutation in only 1 of 6 redundant CaM-encoding alleles, we devised a strategy using CRISPR interference to selectively suppress the mutant gene while sparing the wild-type counterparts. Indeed, suppression of CALM2 expression produced a functional rescue in induced pluripotent stem cell-derived cardiomyocytes with D130G-CALM2, as shown by the normalization of action potential duration and Ca2+/CaM-dependent inactivation after treatment. Moreover, CRISPR interference can be designed to achieve selective knockdown of any of the 3 CALM genes, making it a generalizable therapeutic strategy for any calmodulinopathy. CONCLUSIONS: Overall, this therapeutic strategy holds great promise for calmodulinopathy patients as it represents a generalizable intervention capable of specifically altering CaM expression and potentially attenuating LQTS-triggered cardiac events, thus initiating a path toward precision medicine.


Asunto(s)
Calmodulina/genética , Células Madre Pluripotentes Inducidas/fisiología , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/terapia , Medicina de Precisión/métodos , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Síndrome de QT Prolongado/diagnóstico , Mutación Missense/genética
13.
Nat Commun ; 7: 13709, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27922011

RESUMEN

The stoichiometry of macromolecular interactions is fundamental to cellular signalling yet challenging to detect from living cells. Fluorescence resonance energy transfer (FRET) is a powerful phenomenon for characterizing close-range interactions whereby a donor fluorophore transfers energy to a closely juxtaposed acceptor. Recognizing that FRET measured from the acceptor's perspective reports a related but distinct quantity versus the donor, we utilize the ratiometric comparison of the two to obtain the stoichiometry of a complex. Applying this principle to the long-standing controversy of calmodulin binding to ion channels, we find a surprising Ca2+-induced switch in calmodulin stoichiometry with Ca2+ channels-one calmodulin binds at basal cytosolic Ca2+ levels while two calmodulins interact following Ca2+ elevation. This feature is curiously absent for the related Na channels, also potently regulated by calmodulin. Overall, our assay adds to a burgeoning toolkit to pursue quantitative biochemistry of dynamic signalling complexes in living cells.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Sustancias Macromoleculares/metabolismo , Animales , Canales de Calcio/metabolismo , Calmodulina/metabolismo , Supervivencia Celular , Células HEK293 , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Miosina Tipo V/química , Miosina Tipo V/metabolismo , Unión Proteica , Dominios Proteicos , Reproducibilidad de los Resultados , Canales de Sodio/metabolismo
14.
Nat Protoc ; 11(12): 2470-2498, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27831569

RESUMEN

Förster resonance energy transfer (FRET) is a versatile method for analyzing protein-protein interactions within living cells. This protocol describes a nondestructive live-cell FRET assay for robust quantification of relative binding affinities for protein-protein interactions. Unlike other approaches, our method correlates the measured FRET efficiencies to relative concentration of interacting proteins to determine binding isotherms while including collisional FRET corrections. We detail how to assemble and calibrate the equipment using experimental and theoretical procedures. A step-by-step protocol is given for sample preparation, data acquisition and analysis. The method uses relatively inexpensive and widely available equipment and can be performed with minimal training. Implementation of the imaging setup requires up to 1 week, and sample preparation takes ∼1-3 d. An individual FRET experiment, including control measurements, can be completed within 4-6 h, with data analysis requiring an additional 1-3 h.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Técnicas del Sistema de Dos Híbridos , Supervivencia Celular , Transferencia Resonante de Energía de Fluorescencia/instrumentación , Células HEK293 , Humanos , Técnicas del Sistema de Dos Híbridos/instrumentación
15.
Nat Commun ; 7: 12239, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27456671

RESUMEN

The regulation of L-type Ca(2+) channels by protein kinase A (PKA) represents a crucial element within cardiac, skeletal muscle and neurological systems. Although much work has been done to understand this regulation in cardiac CaV1.2 Ca(2+) channels, relatively little is known about the closely related CaV1.4 L-type Ca(2+) channels, which feature prominently in the visual system. Here we find that CaV1.4 channels are indeed modulated by PKA phosphorylation within the inhibitor of Ca(2+)-dependent inactivation (ICDI) motif. Phosphorylation of this region promotes the occupancy of calmodulin on the channel, thus increasing channel open probability (PO) and Ca(2+)-dependent inactivation. Although this interaction seems specific to CaV1.4 channels, introduction of ICDI1.4 to CaV1.3 or CaV1.2 channels endows these channels with a form of PKA modulation, previously unobserved in heterologous systems. Thus, this mechanism may not only play an important role in the visual system but may be generalizable across the L-type channel family.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Secuencia de Aminoácidos , Animales , Canales de Calcio Tipo L/química , Activación Enzimática , Cobayas , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Dominios Proteicos , Ingeniería de Proteínas , Ratas , Proteínas Recombinantes/metabolismo
16.
Sci Rep ; 6: 27235, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27255217

RESUMEN

CaV1.3 channels are a major class of L-type Ca(2+) channels which contribute to the rhythmicity of the heart and brain. In the brain, these channels are vital for excitation-transcription coupling, synaptic plasticity, and neuronal firing. Moreover, disruption of CaV1.3 function has been associated with several neurological disorders. Here, we focus on the de novo missense mutation A760G which has been linked to autism spectrum disorder (ASD). To explore the role of this mutation in ASD pathogenesis, we examined the effects of A760G on CaV1.3 channel gating and regulation. Introduction of the mutation severely diminished the Ca(2+)-dependent inactivation (CDI) of CaV1.3 channels, an important feedback system required for Ca(2+) homeostasis. This reduction in CDI was observed in two major channel splice variants, though to different extents. Using an allosteric model of channel gating, we found that the underlying mechanism of CDI reduction is likely due to enhanced channel opening within the Ca(2+)-inactivated mode. Remarkably, the A760G mutation also caused an opposite increase in voltage-dependent inactivation (VDI), resulting in a multifaceted mechanism underlying ASD. When combined, these regulatory deficits appear to increase the intracellular Ca(2+) concentration, thus potentially disrupting neuronal development and synapse formation, ultimately leading to ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Canales de Calcio Tipo L/genética , Canales de Calcio/genética , Calcio/metabolismo , Mutación Missense , Animales , Trastorno del Espectro Autista/metabolismo , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Regulación hacia Abajo , Células HEK293 , Homeostasis , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Modelos Animales , Ratas
17.
Cell Rep ; 14(12): 3019-29, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26997269

RESUMEN

Biology has been revolutionized by tools that allow the detection and characterization of protein-protein interactions (PPIs). Förster resonance energy transfer (FRET)-based methods have become particularly attractive as they allow quantitative studies of PPIs within the convenient and relevant context of living cells. We describe here an approach that allows the rapid construction of live-cell FRET-based binding curves using a commercially available flow cytometer. We illustrate a simple method for absolutely calibrating the cytometer, validating our binding assay against the gold standard isothermal calorimetry (ITC), and using flow cytometric FRET to uncover the structural and functional effects of the Cushing-syndrome-causing mutation (L206R) on PKA's catalytic subunit. We discover that this mutation not only differentially affects PKAcat's binding to its multiple partners but also impacts its rate of catalysis. These findings improve our mechanistic understanding of this disease-causing mutation, while illustrating the simplicity, general applicability, and power of flow cytometric FRET.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Calorimetría , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Mapas de Interacción de Proteínas
18.
Nat Commun ; 7: 10370, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26822303

RESUMEN

Timothy Syndrome (TS) is a multisystem disorder, prominently featuring cardiac action potential prolongation with paroxysms of life-threatening arrhythmias. The underlying defect is a single de novo missense mutation in CaV1.2 channels, either G406R or G402S. Notably, these mutations are often viewed as equivalent, as they produce comparable defects in voltage-dependent inactivation and cause similar manifestations in patients. Yet, their effects on calcium-dependent inactivation (CDI) have remained uncertain. Here, we find a significant defect in CDI in TS channels, and uncover a remarkable divergence in the underlying mechanism for G406R versus G402S variants. Moreover, expression of these TS channels in cultured adult guinea pig myocytes, combined with a quantitative ventricular myocyte model, reveals a threshold behaviour in the induction of arrhythmias due to TS channel expression, suggesting an important therapeutic principle: a small shift in the complement of mutant versus wild-type channels may confer significant clinical improvement.


Asunto(s)
Arritmias Cardíacas/metabolismo , Trastorno Autístico/metabolismo , Calcio/metabolismo , Síndrome de QT Prolongado/metabolismo , Sindactilia/metabolismo , Animales , Arritmias Cardíacas/genética , Trastorno Autístico/genética , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Femenino , Cobayas , Humanos , Síndrome de QT Prolongado/genética , Masculino , Mutación Missense , Miocitos Cardíacos/metabolismo , Sindactilia/genética
19.
J Physiol ; 593(17): 3865-84, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26096996

RESUMEN

Novel fluorescence resonance energy transfer-based genetically encoded reporters of calcineurin are constructed by fusing the two subunits of calcineurin with P2A-based linkers retaining the expected native conformation of calcineurin. Calcineurin reporters display robust responses to calcium transients in HEK293 cells. The sensor responses are correlated with NFATc1 translocation dynamics in HEK293 cells. The sensors are uniformly distributed in neonatal myocytes and respond efficiently to single electrically evoked calcium transients and show cumulative activation at frequencies of 0.5 and 1 Hz. In adult myocytes, the calcineurin sensors appear to be localized to the cardiac z-lines, and respond to cumulative calcium transients at frequencies of 0.5 and 1 Hz. The phosphatase calcineurin is a central component of many calcium signalling pathways, relaying calcium signals from the plasma membrane to the nucleus. It has critical functions in a multitude of systems, including immune, cardiac and neuronal. Given the widespread importance of calcineurin in both normal and pathological conditions, new tools that elucidate the spatiotemporal dynamics of calcineurin activity would be invaluable. Here we develop two separate genetically encoded fluorescence resonance energy transfer (FRET)-based sensors of calcineurin activation, DuoCaN and UniCaN. Both sensors showcase a large dynamic range and rapid response kinetics, differing primarily in the linker structure between the FRET pairs. Both sensors were calibrated in HEK293 cells and their responses correlated well with NFAT translocation to the nucleus, validating the biological relevance of the sensor readout. The sensors were subsequently expressed in neonatal rat ventricular myocytes and acutely isolated adult guinea pig ventricular myocytes. Both sensors demonstrated robust responses in myocytes and revealed kinetic differences in calcineurin activation during changes in pacing rate for neonatal versus adult myocytes. Finally, mathematical modelling combined with quantitative FRET measurements provided novel insights into the kinetics and integration of calcineurin activation in response to myocyte Ca transients. In all, DuoCaN and UniCaN stand as valuable new tools for understanding the role of calcineurin in normal and pathological signalling.


Asunto(s)
Calcineurina/fisiología , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Transferencia Resonante de Energía de Fluorescencia , Cobayas , Células HEK293 , Humanos , Factores de Transcripción NFATC/fisiología , Ratas
20.
Curr Mol Pharmacol ; 8(2): 188-205, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25966688

RESUMEN

Voltage-gated Na and Ca(2+) channels represent two major ion channel families that enable myriad biological functions including the generation of action potentials and the coupling of electrical and chemical signaling in cells. Calmodulin regulation (calmodulation) of these ion channels comprises a vital feedback mechanism with distinct physiological implications. Though long-sought, a shared understanding of the channel families remained elusive for two decades as the functional manifestations and the structural underpinnings of this modulation often appeared to diverge. Here, we review recent advancements in the understanding of calmodulation of Ca(2+) and Na channels that suggest a remarkable similarity in their regulatory scheme. This interrelation between the two channel families now paves the way towards a unified mechanistic framework to understand vital calmodulin-dependent feedback and offers shared principles to approach related channelopathic diseases. An exciting era of synergistic study now looms.


Asunto(s)
Canales de Calcio/metabolismo , Calmodulina/metabolismo , Retroalimentación Fisiológica/fisiología , Activación del Canal Iónico/fisiología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Calcio/metabolismo , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...