Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
CA Cancer J Clin ; 73(6): 597-619, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37490348

RESUMEN

Salivary gland cancers are a rare, histologically diverse group of tumors. They range from indolent to aggressive and can cause significant morbidity and mortality. Surgical resection remains the mainstay of treatment, but radiation and systemic therapy are also critical parts of the care paradigm. Given the rarity and heterogeneity of these cancers, they are best managed in a multidisciplinary program. In this review, the authors highlight standards of care as well as exciting new research for salivary gland cancers that will strive for better patient outcomes.


Asunto(s)
Neoplasias de las Glándulas Salivales , Humanos , Neoplasias de las Glándulas Salivales/diagnóstico , Neoplasias de las Glándulas Salivales/terapia
2.
Cancer Immunol Immunother ; 73(1): 10, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38231444

RESUMEN

The antigen processing machinery (APM) components needed for a tumor cell to present an antigen to a T cell are expressed at low levels in solid tumors, constituting an important mechanism of immune escape. More than most other solid tumors, head and neck squamous cell carcinoma (HNSCC) cells tend to have low APM expression, rendering them insensitive to immune checkpoint blockade and most other forms of immunotherapy. In HNSCC, this APM deficiency is largely driven by high levels of EGFR and SHP2, leading to low expression and activation of STAT1; however, recent studies suggest that p53, which is often mutated in HNSCCs, may also play a role. In the current study, we aimed to investigate the extent to which STAT1 and p53 individually regulate APM component expression in HNSCC cells. We found that in cells lacking functional p53, APM expression could still be induced by interferon-gamma or DNA-damaging chemotherapy (cisplatin) as long as STAT1 expression remained intact; when both transcription factors were knocked down, APM component expression was abolished. When we bypassed these deficient pathways by rescuing the expression of NLRC5, APM expression was also restored. These results suggest that dual loss of functional STAT1 and p53 may render HNSCC cells incapable of processing and presenting antigens, but rescue of downstream NLRC5 expression may be an attractive strategy for restoring sensitivity to T cell-based immunotherapy.


Asunto(s)
Presentación de Antígeno , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Proteína p53 Supresora de Tumor/genética , Neoplasias de Cabeza y Cuello/genética , Cisplatino , Factor de Transcripción STAT1/genética , Péptidos y Proteínas de Señalización Intracelular
3.
Artículo en Inglés | MEDLINE | ID: mdl-36538901

RESUMEN

OBJECTIVES: The aim of the study was to investigate the association of surgical margin conditions, including positive specimen margins revised to negative relative to local recurrence, disease-free survival, and overall survival (OS) within a cohort of HPV-mediated oropharyngeal squamous cell carcinoma (OPSCC) who underwent en bloc resection via transoral robotic surgery (TORS). MATERIALS AND METHODS: Retrospective cohort of patients with untreated HPV-mediated OPSCC cT1 or T2 undergoing TORS resection between October 2014 and March 2020. The methodologic description of our interdisciplinary institutional approach, number of cut-through margins (CTMs) during intraoperative consultation, percentage of final positive margin cases, and disease-free survival and OS stratified by margin status and margin tumor-free distance is identified. RESULTS: 135 patients with primary cT1/T2 HPV-mediated OPSCC met inclusion criteria. Twenty-eight of 135 (20.7%) specimens revealed CTM and were revised during the same operative setting. Three of 135 (2.2%) surgical cases had positive final margin status. Local control rate was 97%. On univariate analysis, margin distance did not impact OS. CTM and final positive margins had lower OS than initially negative margins (p = 0.044). Pathologic N-stage significantly impacted OS (p < 0.001). CONCLUSIONS: High local control rate and low final positive margin status confound the study of specimen margin-based techniques in HPV-mediated OPSCC resected en bloc with TORS. Pathologic N-stage may impact OS more than margin status. Larger numbers are needed to confirm differences.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias Orofaríngeas , Infecciones por Papillomavirus , Procedimientos Quirúrgicos Robotizados , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/cirugía , Márgenes de Escisión , Carcinoma de Células Escamosas/cirugía , Carcinoma de Células Escamosas/patología , Neoplasias Orofaríngeas/cirugía , Neoplasias Orofaríngeas/patología , Procedimientos Quirúrgicos Robotizados/métodos , Estudios Retrospectivos , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/cirugía
4.
Ann Surg Oncol ; 29(11): 7002-7006, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35842530

RESUMEN

BACKGROUND: Delays in care can lead to inferior survival outcomes in head and neck cancer and other cancers. In the case of malignancies for which surgery is the preferred primary treatment modality, challenges in surgical scheduling can present a major hurdle to initiating definitive therapy in a timely fashion. It is critical to maintain efficient use of operating room resources. Traditionally, surgery is scheduled with the surgeon who initially saw the patient in consultation, and timing of surgery is tightly linked to the availability and operating room block time of the individual surgeon. METHODS: Scheduling of oncologic head and neck surgery was transitioned from a surgeon-specific method to a team-based approach wherein a patient in need of oncologic head and neck surgery is scheduled with the next-available surgeon with appropriate expertise. RESULTS: Despite substantial growth of our practice, transition to a team-based scheduling approach allowed us to maintain high utilization of operating room block time. Patient and surgeon satisfaction remain high with this new system. CONCLUSIONS: A team-based surgical scheduling approach can help optimize operating room utilization and minimize delays in cancer care, potentially leading to improved oncologic outcomes.


Asunto(s)
Neoplasias de Cabeza y Cuello , Cirujanos , Citas y Horarios , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Quirófanos , Derivación y Consulta
5.
Am J Otolaryngol ; 43(3): 103440, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35398743

RESUMEN

BACKGROUND: A total glossectomy (TG) may be required for advanced tongue tumors. TG with total laryngectomy (TGL) may be indicated in some cases with tumor extension into the larynx or high risk of aspiration. Total glossectomy with laryngeal preservation (TGLP) may preserve phonation ability relative to TGL, yet TGLP may increase the risk of aspiration. METHODS: For this narrative review, we performed a comprehensive literature search of studies relevant to TG and TGL. Clinical studies investigating survival, functional outcomes, and quality of life in following TGLP or TGL were of particular interest. RESULTS: Few studies in the literature directly compare survival, functional, and quality of life (QOL) outcomes between TGLP and TGL. TGLP is associated with intelligible speech. However, studies investigating gastrostomy tube dependence following TGLP versus TGL have generated conflicting results. CONCLUSION: Further research on functional and QOL outcomes in patients undergoing TGL or TGLP is needed.


Asunto(s)
Laringe , Neoplasias de la Lengua , Glosectomía/métodos , Humanos , Laringectomía , Calidad de Vida , Estudios Retrospectivos , Neoplasias de la Lengua/cirugía
6.
Physiol Rev ; 94(2): 609-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24692356

RESUMEN

About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.


Asunto(s)
Arritmias Cardíacas/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Miocardio/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Potenciales de Acción , Animales , Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/fisiopatología , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Periodo Refractario Electrofisiológico
7.
FASEB J ; 33(2): 2537-2552, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30289750

RESUMEN

KCNE5 is an X-linked gene encoding KCNE5, an ancillary subunit to voltage-gated potassium (KV) channels. Human KCNE5 mutations are associated with atrial fibrillation (AF)- and Brugada syndrome (BrS)-induced cardiac arrhythmias that can arise from increased potassium current in cardiomyocytes. Seeking to establish underlying molecular mechanisms, we created and studied Kcne5 knockout ( Kcne5-/0) mice. Intracardiac ECG revealed that Kcne5 deletion caused ventricular premature beats, increased susceptibility to induction of polymorphic ventricular tachycardia (60 vs. 24% in Kcne5+/0 mice), and 10% shorter ventricular refractory period. Kcne5 deletion increased mean ventricular myocyte KV current density in the apex and also in the subpopulation of septal myocytes that lack fast transient outward current ( Ito,f). The current increases arose from an apex-specific increase in slow transient outward current-1 ( IKslow,1) (conducted by KV1.5) and Ito,f (conducted by KV4) and an increase in IKslow,2 (conducted by KV2.1) in both apex and septum. Kcne5 protein localized to the intercalated discs in ventricular myocytes, where KV2.1 was also detected in both Kcne5-/0 and Kcne5+/0 mice. In HL-1 cardiac cells and human embryonic kidney cells, KCNE5 and KV2.1 colocalized at the cell surface, but predominantly in intracellular vesicles, suggesting that Kcne5 deletion increases IK,slow2 by reducing KV2.1 intracellular sequestration. The human AF-associated mutation KCNE5-L65F negative shifted the voltage dependence of KV2.1-KCNE5 channels, increasing their maximum current density >2-fold, whereas BrS-associated KCNE5 mutations produced more subtle negative shifts in KV2.1 voltage dependence. The findings represent the first reported native role for Kcne5 and the first demonstrated Kcne regulation of KV2.1 in mouse heart. Increased KV current is a manifestation of KCNE5 disruption that is most likely common to both mouse and human hearts, providing a plausible mechanistic basis for human KCNE5-linked AF and BrS.-David, J.-P., Lisewski, U., Crump, S. M., Jepps, T. A., Bocksteins, E., Wilck, N., Lossie, J., Roepke, T. K., Schmitt, N., Abbott, G. W. Deletion in mice of X-linked, Brugada syndrome- and atrial fibrillation-associated Kcne5 augments ventricular KV currents and predisposes to ventricular arrhythmia.


Asunto(s)
Fibrilación Atrial/complicaciones , Síndrome de Brugada/complicaciones , Genes Ligados a X , Activación del Canal Iónico , Miocitos Cardíacos/patología , Canales de Potasio con Entrada de Voltaje/fisiología , Taquicardia Ventricular/etiología , Animales , Fibrilación Atrial/genética , Síndrome de Brugada/genética , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Eliminación de Secuencia , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología
8.
Pacing Clin Electrophysiol ; 43(2): 210-216, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31899541

RESUMEN

BACKGROUND: The slow delayed rectifier potassium current IKs is crucial for the repolarization of the cardiac action potential. It is conducted by the voltage-gated channel KV 7.1 encoded by KCNQ1, together with its ß-subunit KCNE1. Loss-of-function (LOF) mutations in KCNQ1 have been associated with heritable cardiac arrhythmias such as Long QT syndrome (LQTS). This disease is characterized by prolonged ventricular repolarization and propensity to ventricular tachyarrhythmia that may lead to syncope, cardiac arrest, and sudden death. We aimed to functionally characterize two KV 7.1 mutations (p.A150T and p.L374H) identified in two independent LQTS patients with different severity of disease phenotype, family history, and co-segregation of LQTS. METHODS: We performed whole-cell patch clamp recordings in CHO-K1 cells, and confocal imaging in Madin-Darby Canine Kidney (MDCK) cells. RESULTS: IKs -A150T showed significantly decreased current amplitudes from above +20 mV (approximately 52% decrease at +40 mV), but demonstrated cell membrane localization similar to wild-type (WT). IKs -L374H, however, exhibited a complete LOF compared to WT channels. Confocal imaging showed endoplasmic reticulum retention of the channel in MDCK cells. Mimicking the heterozygous state of the patients by co-expressing WT and mutant subunits resulted in an approximately 22% decrease in current at +40 mV for A150T. The L374H mutation showed a more pronounced effect (62% reduction at +40 mV compared to WT channel). CONCLUSION: Both mutations, KV 7.1 A150T and L374H, led to loss of channel function. The degree of LOF may mirror the disease phenotype observed in the patients.


Asunto(s)
Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Canales de Potasio con Entrada de Voltaje/genética , Potenciales de Acción , Adulto , Animales , Células CHO , Cricetulus , Perros , Humanos , Mutación con Pérdida de Función , Células de Riñón Canino Madin Darby , Persona de Mediana Edad , Técnicas de Placa-Clamp , Linaje , Fenotipo
9.
J Neurosci ; 37(48): 11523-11536, 2017 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-29042434

RESUMEN

Kv2.1 is a major delayed-rectifier voltage-gated potassium channel widely expressed in neurons of the CNS. Kv2.1 localizes in high-density cell-surface clusters in the soma and proximal dendrites as well as in the axon initial segment (AIS). Given the crucial roles of both of these compartments in integrating signal input and then generating output, this localization of Kv2.1 is ideal for regulating the overall excitability of neurons. Here we used fluorescence recovery after photobleaching imaging, mutagenesis, and pharmacological interventions to investigate the molecular mechanisms that control the localization of Kv2.1 in these two different membrane compartments in cultured rat hippocampal neurons of mixed sex. Our data uncover a unique ability of Kv2.1 channels to use two molecularly distinct trafficking pathways to accomplish this. Somatodendritic Kv2.1 channels are targeted by the conventional secretory pathway, whereas axonal Kv2.1 channels are targeted by a nonconventional trafficking pathway independent of the Golgi apparatus. We further identified a new AIS trafficking motif in the C-terminus of Kv2.1, and show that putative phosphorylation sites in this region are critical for the restricted and clustered localization in the AIS. These results indicate that neurons can regulate the expression and clustering of Kv2.1 in different membrane domains independently by using two distinct localization mechanisms, which would allow neurons to precisely control local membrane excitability.SIGNIFICANCE STATEMENT Our study uncovered a novel mechanism that targets the Kv2.1 voltage-gated potassium channel to two distinct trafficking pathways and two distinct subcellular destinations: the somatodendritic plasma membrane and that of the axon initial segment. We also identified a distinct motif, including putative phosphorylation sites, that is important for the AIS localization. This raises the possibility that the destination of a channel protein can be dynamically regulated via changes in post-translational modification, which would impact the excitability of specific membrane compartments.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Vías Secretoras/fisiología , Canales de Potasio Shab/metabolismo , Animales , Segmento Inicial del Axón/química , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Femenino , Células HEK293 , Hipocampo/química , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Masculino , Neuronas/química , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Ratas , Canales de Potasio Shab/análisis
10.
J Physiol ; 596(20): 4983-4994, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30079574

RESUMEN

KEY POINTS: GABA is an essential molecule for sensory information processing. It is usually assumed to be released by neurons. Here we show that in the dorsal horn of the spinal cord, astrocytes respond to glutamate by releasing GABA. Our findings suggest a novel role for astrocytes in somatosensory information processing. ABSTRACT: Astrocytes participate in neuronal signalling by releasing gliotransmitters in response to neurotransmitters. We investigated if astrocytes from the dorsal horn of the spinal cord of adult red-eared turtles (Trachemys scripta elegans) release GABA in response to glutamatergic receptor activation. For this, we developed a GABA sensor consisting of HEK cells expressing GABAA receptors. By positioning the sensor recorded in the whole-cell patch-clamp configuration within the dorsal horn of a spinal cord slice, we could detect GABA in the extracellular space. Puff application of glutamate induced GABA release events with time courses that exceeded the duration of inhibitory postsynaptic currents by one order of magnitude. Because the events were neither affected by extracellular addition of nickel, cadmium and tetrodotoxin nor by removal of Ca2+ , we concluded that they originated from non-neuronal cells. Immunohistochemical staining allowed the detection of GABA in a fraction of dorsal horn astrocytes. The selective stimulation of A∂ and C fibres in a dorsal root filament induced a Ca2+ increase in astrocytes loaded with Oregon Green BAPTA. Finally, chelating Ca2+ in a single astrocyte was sufficient to prevent the GABA release evoked by glutamate. Our results indicate that glutamate triggers the release of GABA from dorsal horn astrocytes with a time course compatible with the integration of sensory inputs.


Asunto(s)
Astrocitos/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Potenciales Sinápticos , Ácido gamma-Aminobutírico/metabolismo , Animales , Calcio/metabolismo , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Neuronas/metabolismo , Neuronas/fisiología , Asta Dorsal de la Médula Espinal/citología , Asta Dorsal de la Médula Espinal/fisiología , Tortugas
11.
Cell Physiol Biochem ; 47(4): 1742-1750, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29949809

RESUMEN

BACKGROUND/AIMS: The voltage-gated potassium channel KV11.1 has been originally cloned from the brain and is expressed in a variety of tissues. The role of phosphorylation for channel function is a matter of debate. In this study, we aimed to elucidate the extent and role of protein kinase D mediated phosphorylation. METHODS: We employed mass spectrometry, whole-cell patch clamp electrophysiology, confocal microscopy, site-directed mutagenesis, and western blotting. RESULTS: Using brain tissue from rat and mouse, we mapped several phosphorylated KV11.1 residues by LC-MS mass spectrometry and identified protein kinase D (PKD1) as possible regulatory kinase. Co-expression of KV11.1 with PKD1 reduced current amplitudes without altering protein levels or surface expression of the channel. Based on LC-MS results from in vivo and HEK293 cell experiments we chose four KV11.1 mutant candidates for further functional analysis. Ablation of the putative PKD phosphorylation site in the mutant S284A increased the maximal current indicating S284 as a main PKD target in KV11.1. CONCLUSIONS: Our data might help mitigating a long-standing controversy in the field regarding PKC regulation of KV11.1. We propose that PKD1 mediates the PKC effects on KV11.1 and we found that PKD targets S284 in the N-terminus of the channel.


Asunto(s)
Encéfalo/metabolismo , Canal de Potasio ERG1/metabolismo , Proteína Quinasa C/metabolismo , Sustitución de Aminoácidos , Animales , Canal de Potasio ERG1/genética , Células HEK293 , Humanos , Ratones , Mutación Missense , Fosforilación/genética , Proteína Quinasa C/genética , Ratas
12.
PLoS Comput Biol ; 13(6): e1005587, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28622331

RESUMEN

A recent experimental study investigating patients with lone atrial fibrillation identified six novel mutations in the KCNA5 gene. The mutants exhibited both gain- and loss-of-function of the atrial specific ultra-rapid delayed rectifier K+ current, IKur. The aim of this study is to elucidate and quantify the functional impact of these KCNA5 mutations on atrial electrical activity. A multi-scale model of the human atria was updated to incorporate detailed experimental data on IKur from both wild-type and mutants. The effects of the mutations on human atrial action potential and rate dependence were investigated at the cellular level. In tissue, we assessed the effects of the mutations on the vulnerability to unidirectional conduction patterns and dynamics of re-entrant excitation waves. Gain-of-function mutations shortened the action potential duration in single cells, and stabilised and accelerated re-entrant excitation in tissue. Loss-of-function mutations had heterogeneous effects on action potential duration and promoted early-after-depolarisations following beta-adrenergic stimulation. In the tissue model, loss-of-function mutations facilitated breakdown of excitation waves at more physiological excitation rates than the wild-type, and the generation of early-after-depolarisations promoted unidirectional patterns of excitation. Gain- and loss-of-function IKur mutations produced multiple mechanisms of atrial arrhythmogenesis, with significant differences between the two groups of mutations. This study provides new insights into understanding the mechanisms by which mutant IKur contributes to atrial arrhythmias. In addition, as IKur is an atrial-specific channel and a number of IKur-selective blockers have been developed as anti-AF agents, this study also helps to understand some contradictory results on both pro- and anti-arrhythmic effects of blocking IKur.


Asunto(s)
Fibrilación Atrial/genética , Variación Genética/genética , Sistema de Conducción Cardíaco/fisiopatología , Canal de Potasio Kv1.5/genética , Modelos Cardiovasculares , Modelos Genéticos , Simulación por Computador , Humanos , Activación del Canal Iónico/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal/genética , Relación Estructura-Actividad
13.
Pacing Clin Electrophysiol ; 41(6): 620-626, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29572929

RESUMEN

BACKGROUND: Dysfunction of NaV 1.5 encoded by SCN5A accounts for approximately half of the channelopathic SIDS cases. We investigated the functional effect of two gene variants identified in the same patient, one in SCN5A and one in SCN1Bb. The aim of the study was to risk stratify the proband's family. METHODS: The family was referred for cardiovascular genetic evaluation to assess familial risk of cardiac disease. Functional analysis of the identified variants was performed with patch-clamp electrophysiology in HEK293 cells. RESULTS: A 16-month-old healthy boy died suddenly in the context of nonspecific illness and possible fever. Postmortem genetic testing revealed variants in the SCN5A and SCN1Bb genes. The proband's father carries the same variants but is asymptomatic. Electrophysiological analysis of the NaV 1.5_1281X truncation revealed complete loss-of-function of the channel. Coexpression of NaV 1.5 with NaV ß1b significantly increased INa density when compared to NaV 1.5 alone. The NaV ß1b _V268I variant abolished this INa density increase. Moreover, it shifted the activation curve toward more depolarized potentials. CONCLUSIONS: Genetic variation of both sodium channel and its modifiers may contribute to sudden unexplained death in childhood. However, the asymptomatic father suggests that genetic variation of these genes is not sufficient to cause sudden death or clinically detectable SCN5A phenotypes.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.5/genética , Muerte Súbita del Lactante/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Variación Genética , Humanos , Lactante , Masculino , Linaje
14.
Proc Natl Acad Sci U S A ; 112(18): 5714-9, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25901329

RESUMEN

Polyunsaturated fatty acids (PUFAs) affect cardiac excitability. Kv7.1 and the ß-subunit KCNE1 form the cardiac IKs channel that is central for cardiac repolarization. In this study, we explore the prospects of PUFAs as IKs channel modulators. We report that PUFAs open Kv7.1 via an electrostatic mechanism. Both the polyunsaturated acyl tail and the negatively charged carboxyl head group are required for PUFAs to open Kv7.1. We further show that KCNE1 coexpression abolishes the PUFA effect on Kv7.1 by promoting PUFA protonation. PUFA analogs with a decreased pKa value, to preserve their negative charge at neutral pH, restore the sensitivity to open IKs channels. PUFA analogs with a positively charged head group inhibit IKs channels. These different PUFA analogs could be developed into drugs to treat cardiac arrhythmias. In support of this possibility, we show that PUFA analogs act antiarrhythmically in embryonic rat cardiomyocytes and in isolated perfused hearts from guinea pig.


Asunto(s)
Antiarrítmicos/metabolismo , Arritmias Cardíacas/tratamiento farmacológico , Ácidos Grasos Insaturados/metabolismo , Canal de Potasio KCNQ1/química , Mutación , Animales , Conductividad Eléctrica , Femenino , Cobayas , Corazón/efectos de los fármacos , Humanos , Canal de Potasio KCNQ1/genética , Microscopía Electrónica de Rastreo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Oocitos/metabolismo , Perfusión , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Electricidad Estática , Xenopus laevis
15.
Int J Audiol ; 57(sup4): S49-S54, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-28728452

RESUMEN

OBJECTIVE: Review of the literature regarding hearing loss in patients with head and neck cancer treated with chemoradiation. DESIGN: Studies in the literature are reviewed that pertain to hearing loss sustained in head and neck cancer patients receiving cisplatin-based chemoradiation. Personal observations noted while treating these patients are also detailed. STUDY SAMPLE: PubMed was searched for pertinent articles regarding hearing loss in head and neck cancer patients receiving cisplatin chemotherapy and/or radiation. RESULTS: Studies on the incidence and severity of hearing loss in head and neck cancer patients are limited, but those studies suggest that the risk of hearing loss is greater with higher-dose regimens. CONCLUSIONS: Newer cisplatin chemotherapy regimens using lower, weekly doses may be associated with a lower incidence and severity of hearing loss; however, large prospective studies are needed. Such information will be paramount to effective pre-treatment counselling of head and neck cancer patients.


Asunto(s)
Antineoplásicos/efectos adversos , Quimioradioterapia/efectos adversos , Cisplatino/efectos adversos , Neoplasias de Cabeza y Cuello/terapia , Pérdida Auditiva/inducido químicamente , Audición/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas/métodos , Neoplasias de Cabeza y Cuello/diagnóstico , Audición/efectos de la radiación , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/prevención & control , Pruebas Auditivas , Humanos , Dosificación Radioterapéutica , Medición de Riesgo , Factores de Riesgo , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico
16.
Cancer ; 123(22): 4382-4390, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28950407

RESUMEN

BACKGROUND: Human papillomavirus 16 (HPV16) E6 antibodies may be an early marker of the diagnosis and recurrence of human papillomavirus-driven oropharyngeal cancer (HPV-OPC). METHODS: This study identified 161 incident oropharyngeal cancer (OPC) cases diagnosed at the University of Pittsburgh (2003-2013) with pretreatment serum. One hundred twelve had preexisting clinical HPV testing with p16 immunohistochemistry and HPV in situ hybridization (87 were dual-positive [HPV-OPC], and 25 were dual-negative [HPV-negative]); 62 had at least 1 posttreatment serum sample. Eighty-six of the 161 tumors were available for additional HPV16 DNA/RNA testing (45 were dual-positive [HPV16-OPC], and 19 were dual-negative [HPV16-negative). HPV16 E6 antibody testing was conducted with multiplex serology. The following were evaluated: 1) the sensitivity and specificity of HPV16 E6 serology for distinguishing HPV-OPC and HPV16-OPC from HPV-negative OPC, 2) HPV16 E6 antibody decay after treatment with linear models accommodating correlations in variance estimates, and 3) pre- and posttreatment HPV16 E6 levels and the risk of recurrence with Cox proportional hazards models. RESULTS: Seventy-eight of 87 HPV-OPCs were HPV16 E6-seropositive (sensitivity, 89.7%; 95% confidence interval [CI], 81.3%-95.2%), and 24 of 25 HPV-negative OPCs were HPV16 E6-seronegative (specificity, 96.0%; 95% CI, 79.6%-99.9%). Forty-two of 45 HPV16-OPCs were HPV16 E6-seropositive (sensitivity, 93.3%; 95% CI, 81.7%-98.6%), and 18 of 19 HPV16-negative OPCs were HPV16 E6-seronegative (specificity, 94.7%; 95% CI, 74.0%-99.9%). Posttreatment HPV16 E6 antibody levels did not decrease significantly from the baseline (P = .575; median follow-up, 307 days) and were not associated with the risk of recurrence. However, pretreatment HPV16 E6 seropositivity was associated with an 86% reduced risk of local/regional recurrence (hazard ratio, 0.14; 95% CI, 0.03-0.68; P = .015). CONCLUSIONS: HPV16 E6 antibodies may have potential clinical utility for the diagnosis and/or prognosis of HPV-OPC. Cancer 2017;123:4382-90. © 2017 American Cancer Society.


Asunto(s)
Anticuerpos Antivirales/sangre , Proteínas Oncogénicas Virales/inmunología , Neoplasias Orofaríngeas/diagnóstico , Infecciones por Papillomavirus/diagnóstico , Proteínas Represoras/inmunología , Transformación Celular Viral/inmunología , Femenino , Papillomavirus Humano 16/inmunología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias Orofaríngeas/sangre , Neoplasias Orofaríngeas/patología , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/sangre , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/patología , Valor Predictivo de las Pruebas , Pronóstico , Recurrencia , Sensibilidad y Especificidad
18.
J Mol Cell Cardiol ; 97: 24-35, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27103460

RESUMEN

The two-pore domain potassium (K(+)) channel TWIK-1 (or K2P1.1) contributes to background K(+) conductance in diverse cell types. TWIK-1, encoded by the KCNK1 gene, is present in the human heart with robust expression in the atria, however its physiological significance is unknown. To evaluate the cardiac effects of TWIK-1 deficiency, we studied zebrafish embryos after knockdown of the two KCNK1 orthologues, kcnk1a and kcnk1b. Knockdown of kcnk1a or kcnk1b individually caused bradycardia and atrial dilation (p<0.001 vs. controls), while ventricular stroke volume was preserved. Combined knockdown of both kcnk1a and kcnk1b resulted in a more severe phenotype, which was partially reversed by co-injection of wild-type human KCNK1 mRNA, but not by a dominant negative variant of human KCNK1 mRNA. To determine whether genetic variants in KCNK1 might cause atrial fibrillation (AF), we sequenced protein-coding regions in two independent cohorts of patients (373 subjects) and identified three non-synonymous variants, p.R171H, p.I198M and p.G236S, that were all located in highly conserved amino acid residues. In transfected mammalian cells, zebrafish and wild-type human TWIK-1 channels had a similar cellular distribution with predominant localization in the endosomal compartment. Two-electrode voltage-clamp experiments using Xenopus oocytes showed that both zebrafish and wild-type human TWIK-1 channels produced K(+) currents that are sensitive to external K(+) concentration as well as acidic pH. There were no effects of the three KCNK1 variants on cellular localization, current amplitude or reversal potential at pH7.4 or pH6. Our data indicate that TWIK-1 has a highly conserved role in cardiac function and is required for normal heart rate and atrial morphology. Despite the functional importance of TWIK-1 in the atrium, genetic variation in KCNK1 is not a common primary cause of human AF.


Asunto(s)
Remodelación Atrial/genética , Estudios de Asociación Genética , Atrios Cardíacos/metabolismo , Frecuencia Cardíaca/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Adulto , Anciano , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Variación Genética , Atrios Cardíacos/anatomía & histología , Atrios Cardíacos/patología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Transporte de Proteínas , Factores de Riesgo , Pez Cebra
19.
Pflugers Arch ; 468(4): 643-54, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26729267

RESUMEN

In isolated human atrial cardiomyocytes, inhibition of K2P3.1 K(+) channels results in action potential (action potential duration (APD)) prolongation. It has therefore been postulated that K2P3.1 (KCNK3), together with K2P9.1 (KCNK9), could represent novel drug targets for the treatment of atrial fibrillation (AF). However, it is unknown whether these findings in isolated cells translate to the whole heart. The purposes of this study were to investigate the expression levels of KCNK3 and KCNK9 in human hearts and two relevant rodent models and determine the antiarrhythmic potential of K2P3.1 inhibition in isolated whole-heart preparations. By quantitative PCR, we found that KCNK3 is predominantly expressed in human atria whereas KCNK9 was not detectable in heart human tissue. No differences were found between patients in AF or sinus rhythm. The expression in guinea pig heart resembled humans whereas rats displayed a more uniform expression of KCNK3 between atria and ventricle. In voltage-clamp experiments, ML365 and A293 were found to be potent and selective inhibitors of K2P3.1, but at pH 7.4, they failed to prolong atrial APD and refractory period (effective refractory period (ERP)) in isolated perfused rat and guinea pig hearts. At pH 7.8, which augments K2P3.1 currents, pharmacological channel inhibition produced a significant prolongation of atrial ERP (11.6 %, p = 0.004) without prolonging ventricular APD but did not display a significant antiarrhythmic effect in our guinea pig AF model (3/8 hearts converted on A293 vs 0/7 hearts in time-matched controls). These results suggest that when K2P3.1 current is augmented, K2P3.1 inhibition leads to atrial-specific prolongation of ERP; however, this ERP prolongation did not translate into significant antiarrhythmic effects in our AF model.


Asunto(s)
Potenciales de Acción , Arritmias Cardíacas/metabolismo , Función Atrial , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Protones , Periodo Refractario Electrofisiológico , Adolescente , Adulto , Animales , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Femenino , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Ratas , Ratas Wistar , Especificidad de la Especie , Función Ventricular
20.
Traffic ; 14(4): 399-411, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23324056

RESUMEN

The voltage-gated potassium channel KV 7.1 is regulated by non-pore forming regulatory KCNE ß-subunits. Together with KCNE1, it forms the slowly activating delayed rectifier potassium current IKs . However, where the subunits assemble and which of the subunits determines localization of the IKs -complex has not been unequivocally resolved yet. We employed trafficking-deficient KV 7.1 and KCNE1 mutants to investigate IKs trafficking using the polarized Madin-Darby Canine Kidney cell line. We find that the assembly happens early in the secretory pathway but provide three lines of evidence that it takes place in a post-endoplasmic reticulum compartment. We demonstrate that KV 7.1 targets the IKs -complex to the basolateral membrane, but that KCNE1 can redirect the complex to the apical membrane upon mutation of critical KV 7.1 basolateral targeting signals. Our data provide a possible explanation to the fact that KV 7.1 can be localized apically or basolaterally in different epithelial tissues and offer a solution to divergent literature results regarding the effect of KCNE subunits on the subcellular localization of KV 7.1/KCNE complexes.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Multimerización de Proteína , Subunidades de Proteína/metabolismo , Animales , Membrana Celular/metabolismo , Perros , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/genética , Células de Riñón Canino Madin Darby , Mutación Missense , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Dominios y Motivos de Interacción de Proteínas , Señales de Clasificación de Proteína/genética , Subunidades de Proteína/genética , Transporte de Proteínas , Vías Secretoras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA