Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 27(7): 2100-2110, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33451980

RESUMEN

PURPOSE: Pancreatic cancer is an aggressive disease associated with a poor 5-year overall survival. Most patients are ineligible for surgery due to late diagnosis and are treated primarily with chemotherapy with very limited success. Pancreatic cancer is relatively insensitive to chemotherapy due to multiple factors, including reduced bioavailability of drugs to tumor cells. One strategy to improve drug efficacy with reduced toxicity is the development of antibody-drug conjugates (ADC), which have now been used successfully to treat both solid and liquid tumors. Here, we evaluate the efficacy of TR1801-ADC, a newly developed ADC composed of a MET antibody conjugated to the highly potent pyrrolobenzodiazepine toxin-linker, tesirine. EXPERIMENTAL DESIGN: We first evaluated MET expression and subcellular localization in pancreatic cancer cell lines, human tumors, and patient-derived xenografts (PDX). We then tested TR1801-ADC efficacy in vitro in pancreatic cancer cell lines. Preclinical evaluation of TR1801-ADC efficacy was conducted on PDXs selected on the basis of their MET expression level. RESULTS: We show that MET is highly expressed and located at the plasma membrane of pancreatic cancer cells. We found that TR1801-ADC induces a specific cytotoxicity in pancreatic cancer cell lines and a profound tumor growth inhibition, even in a gemcitabine-resistant tumor. We also noted synergism between TR1801-ADC and gemcitabine in vitro and an improved response to the combination in vivo. CONCLUSIONS: Together, these results suggest the promise of agents such as TR1801-ADC as a novel approach to the treatment of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Inmunoconjugados/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/inmunología , Animales , Carcinoma Ductal Pancreático/mortalidad , Línea Celular Tumoral , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/mortalidad , Proteínas Proto-Oncogénicas c-met/análisis , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
2.
Cancer Biol Ther ; 21(6): 549-559, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32192391

RESUMEN

c-Met is a well-characterized oncogene that is associated with poor prognosis in many solid tumor types. While responses to c-Met inhibitors have been observed in clinical trials, activity appears to be limited to those with MET gene amplifications or mutations. We developed a c-Met targeted antibody-drug conjugate (ADC) with preclinical activity in the absence of MET gene amplification or mutation, and activity even in the context of moderate protein expression. The ADC utilized a high-affinity c-Met antibody (P3D12), that induced c-Met degradation with minimal activation of c-Met signaling, or mitogenic effect. P3D12 was conjugated to the tubulin inhibitor toxin MMAF via a cleavable linker (vc-MMAF). P3D12-vc-MMAF demonstrated potent in vitro activity in c-Met protein-expressing cell lines regardless of MET gene amplification or mutation status, and retained activity in cell lines with medium-low c-Met protein expression. In contrast, the c-Met tyrosine kinase inhibitor (TKI) PHA-665752 slowed tumor cell growth in vitro only in the context of MET gene amplification or very high protein expression. This differential activity was even more marked in vivo. P3D12-vc-MMAF demonstrated robust inhibition of tumor growth in the MET gene amplified MKN-45 xenograft model, and similar results in H1975, which expresses moderate levels of wild type c-Met without genomic amplification. By comparison, the c-Met TKI, PHA-665752, demonstrated modest tumor growth inhibition in MKN-45, and no inhibition at all in H1975. Taken together, these data suggest that P3D12-vc-MMAF may have a superior clinical profile in treating c-Met positive malignancies in contrast to c-Met pathway inhibitors.


Asunto(s)
Amplificación de Genes , Inmunoconjugados/farmacología , Indoles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/inmunología , Neoplasias Gástricas/tratamiento farmacológico , Sulfonas/farmacología , Animales , Anticuerpos Monoclonales/química , Apoptosis , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Oligopéptidos/química , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Oncol ; 14(1): 54-68, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31736230

RESUMEN

cMet is a well-characterized oncogene that is the target of many drugs including small molecule and biologic pathway inhibitors, and, more recently, antibody-drug conjugates (ADCs). However, the clinical benefit from cMet-targeted therapy has been limited. We developed a novel cMet-targeted 'third-generation' ADC, TR1801-ADC, that was optimized at different levels including specificity, stability, toxin-linker, conjugation site, and in vivo efficacy. Our nonagonistic cMet antibody was site-specifically conjugated to the pyrrolobenzodiazepine (PBD) toxin-linker tesirine and has picomolar activity in cancer cell lines derived from different solid tumors including lung, colorectal, and gastric cancers. The potency of our cMet ADC is independent of MET gene copy number, and its antitumor activity was high not only in high cMet-expressing cell lines but also in medium-to-low cMet cell lines (40 000-90 000 cMet/cell) in which a cMet ADC with tubulin inhibitor payload was considerably less potent. In vivo xenografts with low-medium cMet expression were also very responsive to TR1801-ADC at a single dose, while a cMet ADC using a tubulin inhibitor showed a substantially reduced efficacy. Furthermore, TR1801-ADC had excellent efficacy with significant antitumor activity in 90% of tested patient-derived xenograft models of gastric, colorectal, and head and neck cancers: 7 of 10 gastric models, 4 of 10 colorectal cancer models, and 3 of 10 head and neck cancer models showed complete tumor regression after a single-dose administration. Altogether, TR1801-ADC is a new generation cMet ADC with best-in-class preclinical efficacy and good tolerability in rats.


Asunto(s)
Antineoplásicos/farmacología , Benzodiazepinas/farmacología , Inmunoconjugados/farmacología , Neoplasias/tratamiento farmacológico , Oncogenes/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Pirroles/farmacología , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Neoplasias del Sistema Biliar/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Inmunoconjugados/uso terapéutico , Inmunoconjugados/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/inmunología , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Ratas Sprague-Dawley , Neoplasias Gástricas/metabolismo , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Cell Int ; 14(1): 26, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24645697

RESUMEN

BACKGROUND: Primary and secondary brain cancers are highly treatment resistant, and their marked angiogenesis attracts interest as a potential therapeutic target. Recent observations reveal that the microvascular endothelium of primary high-grade gliomas expresses prostate specific membrane antigen (PSMA). Breast cancers express PSMA and they frequently form secondary brain tumors. Hence we report here our pilot study addressing the feasibility of PSMA targeting in brain and metastatic breast tumors, by examining PSMA levels in all glioma grades (19 patients) and in breast cancer brain metastases (5 patients). METHODS: Tumor specimens were acquired from archival material and normal brain tissues from autopsies. Tissue were stained and probed for PSMA, and the expression levels imaged and quantified using automated hardware and software. PSMA staining intensities of glioma subtypes, breast tumors, and breast tumor brain metastases were compared statistically versus normals. RESULTS: Normal brain microvessels (4 autopsies) did not stain for PSMA, while a small proportion (<5%) of healthy neurons stained, and were surrounded by an intact blood brain barrier. Tumor microvessels of the highly angiogenic grade IV gliomas showed intense PSMA staining which varied between patients and was significantly higher (p < 0.05) than normal brain. Grade I gliomas showed moderate vessel staining, while grade II and III gliomas had no vessel staining, but a few (<2%) of the tumor cells stained. Both primary breast cancer tissues and the associated brain metastases exhibited vascular PSMA staining, although the intensity of staining was generally less for the metastatic lesions. CONCLUSIONS: Our results align with and extend previous data showing PSMA expression in blood vessels of gliomas and breast cancer brain metastases. These results provide a rationale for more comprehensive studies to explore PSMA targeted agents for treating secondary brain tumors with PSMA expressing vasculature. Moreover, given that PSMA participates in angiogenesis, cell signaling, tumor survival, and invasion, characterizing its expression may help guide later investigations of the poorly understood process of low grade glioma progression to glioblastoma.

5.
J Comp Neurol ; 522(5): 1072-101, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24096910

RESUMEN

Early electrical activity and calcium influx regulate crucial aspects of neuronal development. Small-conductance calcium-activated potassium (SK) channels regulate action potential firing and shape calcium influx through feedback regulation in mature neurons. These functions, observed in the adult nervous system, make them ideal candidates to regulate activity- and calcium-dependent processes in neurodevelopment. However, to date little is known about the onset of expression and regions expressing SK channel subunits in the embryonic and postnatal development of the central nervous system (CNS). To allow studies on the contribution of SK channels to different phases of development of single neurons and networks, we have performed a detailed in situ hybridization mapping study, providing comprehensive distribution profiles of all three SK subunits (SK1, SK2, and SK3) in the rat CNS during embryonic and postnatal development. SK channel transcripts are expressed at early stages of prenatal CNS development. The three SK channel subunits display different developmental expression gradients in distinct CNS regions, with time points of expression and up- or downregulation that can be associated with a range of diverse developmental events. Their early expression in embryonic development suggests an involvement of SK channels in the regulation of developmental processes. Additionally, this study shows how the postnatal ontogenetic patterns lead to the adult expression map for each SK channel subunit and how their coexpression in the same regions or neurons varies throughout development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Sistema Nervioso , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Embrión de Mamíferos , Sistema Nervioso/embriología , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética
7.
Cell Cycle ; 10(21): 3731-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045127

RESUMEN

Addition of short (6 to 16 amino acids) peptide sequences to the N-terminus of p110α induces a gain of function. Such sequences include the common Flag, His, and VSV tags as well as random sequences. An N-terminal myristylation signal generally believed to activate p110α by providing a constitutive membrane address is also activating, if myristylation is mutationally abolished. The gain of function seen with N-terminally tagged (NTT) p110α constructs extends to signaling, oncogenic transformation and stimulation of cell growth. The activating effect of N-terminal tags requires a functional Ras-binding domain in p110α. Mutations in that domain (T208D and K227A) abolish the gains of function in oncogenicity and signaling. The dominant negative mutant of Ras, RasN17, interferes with transformation induced by NTT p110α. In contrast, binding to p85 activity is not required for cellular transformation and enhanced signaling by NTT p110α.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/fisiología , Transducción de Señal , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Transformación Celular Neoplásica , Embrión de Pollo , Fosfatidilinositol 3-Quinasa Clase Ia/química , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Subunidades de Proteína/fisiología , Proteínas ras/metabolismo
8.
Curr Top Microbiol Immunol ; 347: 79-104, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20582532

RESUMEN

The catalytic and regulatory subunits of class I phosphoinositide 3-kinase (PI3K) have oncogenic potential. The catalytic subunit p110α and the regulatory subunit p85 undergo cancer-specific gain-of-function mutations that lead to enhanced enzymatic activity, ability to signal constitutively, and oncogenicity. The ß, γ, and δ isoforms of p110 are cell-transforming as overexpressed wild-type proteins. Class I PI3Ks have the unique ability to generate phosphoinositide 3,4,5 trisphosphate (PIP(3)). Class II and class III PI3Ks lack this ability. Genetic and cell biological evidence suggests that PIP(3) is essential for PI3K-mediated oncogenicity, explaining why class II and class III enzymes have not been linked to cancer. Mutational analysis reveals the existence of at least two distinct molecular mechanisms for the gain of function seen with cancer-specific mutations in p110α; one causing independence from upstream receptor tyrosine kinases, the other inducing independence from Ras. An essential component of the oncogenic signal that is initiated by PI3K is the TOR (target of rapamycin) kinase. TOR is an integrator of growth and of metabolic inputs. In complex with the raptor protein (TORC1), it controls cap-dependent translation, and this function is essential for PI3K-initiated oncogenesis.


Asunto(s)
Neoplasias/etiología , Fosfatidilinositol 3-Quinasas/fisiología , Animales , Transformación Celular Neoplásica , Humanos , Isoenzimas/fisiología , Mutación , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética
9.
Mol Cancer Res ; 7(7): 1132-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19584261

RESUMEN

Phosphatidylinositol 3-kinases (PI3K) are divided into three classes, which differ in their substrates and products. Class I generates the inositol phospholipids PI(3)P, PI(3,4)P2, and PI(3,4,5)P3 referred as PIP, PIP2, and PIP3, respectively. Class II produces PIP and PIP2, and class III generates only PIP. Substrate and product differences of the three classes are determined by the activation loops of their catalytic domains. Substitution of the class I activation loop with either class II or III activation loop results in a corresponding change of substrate preference and product restriction. We have evaluated such activation loop substitutions to show that oncogenic activity of class I PI3K is linked to the ability to produce PIP3. We further show that reduction of cellular PIP3 levels by the 5'-phosphatase PIPP interferes with PI3K-induced oncogenic transformation. PIPP also attenuates signaling through Akt and target of rapamycin. Class III PI3K fails to induce oncogenic transformation. Likewise, a constitutively membrane-bound class I PI3K mutant retaining only the protein kinase is unable to induce transformation. We conclude that PIP3 is an essential component of PI3K-mediated oncogenesis and that inability to generate PIP3 abolishes oncogenic potential.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Dominio Catalítico , Línea Celular , Embrión de Pollo , Fibroblastos/metabolismo , Inositol Polifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
10.
Curr Opin Genet Dev ; 19(1): 12-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19185485

RESUMEN

Research on PI 3-kinase (PI3K) is undergoing significant shifts in emphasis. Questions that have been dormant for some time are coming to the forefront, such as the relationship of PTEN to PI3K and the role of AKT in PI3K-driven oncogenesis. Two non-alpha isoforms of Class I PI3K are now established as important determinants in cancer: p110beta and p110delta. The oncogenic activities of p110beta include a non-catalytic function, a finding that will have immediate consequences for drug development.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Transformación Celular Neoplásica/genética , Humanos , Modelos Biológicos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética
11.
J Biol Chem ; 283(52): 36234-40, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18927085

RESUMEN

Ether-á-go-go-1 (Eag1) is a CNS-localized voltage-gated potassium channel that is found ectopically expressed in a majority of extracranial solid tumors. While circumstantial evidence linking Eag1 to tumor biology has been well established, the mechanisms by which the channel contributes to tumor progression remain elusive. In this study, we have used in vivo and in vitro techniques to identify a candidate mechanism. A mutation that eliminates ion permeation fails to completely abolish xenograft tumor formation by transfected cells, indicating that Eag1 contributes to tumor progression independently of its primary function as an ion channel. Our data suggest that Eag1 interferes with the cellular mechanism for maintaining oxygen homeostasis, increasing HIF-1 activity, and thereby VEGF secretion and tumor vascularization.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/biosíntesis , Canales de Potasio Éter-A-Go-Go/fisiología , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Femenino , Homeostasis , Humanos , Hipoxia , Ratones , Ratones SCID , Células 3T3 NIH
12.
Methods Enzymol ; 438: 291-305, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18413256

RESUMEN

Signaling by class I phosphatidylinositol 3-kinase (PI3K) controls cell growth, replication, motility, and metabolism. The PI3K pathway commonly shows gain of function in cancer. Two small GTPases, Rheb (Ras homolog enriched in brain) and Ras (rat sarcoma viral oncogene), play important roles in PI3K signaling. Rheb activates the TOR (target of rapamycin) kinase in a GTP-dependent manner; it links TOR to upstream signaling components, including the tuberous sclerosis complex (TSC) and Akt (homolog of the Akt8 murine lymphoma viral oncoprotein). Constitutively active, GTP-bound Rheb is oncogenic in cell culture, and activity that requires farnesylation. Ras activates PI3K by recruitment to the plasma membrane and possibly by inducing a conformational change in the catalytic subunit p110 of PI3K. In return, Ras signaling through the MAP kinase (MAPK) pathway is activated by PIP(3), the product of PI3K. Loss of Ras function can interfere with PI3K signaling. Various lines of evidence suggest complementary roles for PI3K and MAPK signaling in oncogenesis.


Asunto(s)
Neoplasias/fisiopatología , Fosfatidilinositol 3-Quinasas/genética , Animales , Transformación Celular Neoplásica , Fosfatidilinositol 3-Quinasa Clase I , Ratones , Proteínas de Unión al GTP Monoméricas/fisiología , Neoplasias/genética , Neuropéptidos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosfatos de Fosfatidilinositol/fisiología , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal
13.
Trends Biochem Sci ; 32(7): 342-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17561399

RESUMEN

Cancer-specific mutations in the catalytic subunit of phosphatidylinositol 3-kinase (PI3K) p110 alpha occur in diverse tumors in frequencies that can exceed 30%. The majority of these mutations map to one of three hot spots in the gene, and the rest are distributed over much of the PI3K coding sequence. Most of the cancer-specific mutations induce a gain of function that results in oncogenicity, elevated lipid kinase activity and constitutive signaling through the kinases Akt and TOR. The location of the mutations on a model structure of p110 alpha indicates several distinct mechanisms for the gain of function. The mutated p110 alpha proteins are promising cancer targets. Although identification of mutant-specific small-molecule inhibitors seems technically challenging, the therapeutic benefits from such inhibitors could be extremely important.


Asunto(s)
Mutación/genética , Neoplasias/enzimología , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Humanos , Isoenzimas/metabolismo , Modelos Biológicos , Neoplasias/terapia , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/clasificación
14.
Proc Natl Acad Sci U S A ; 104(13): 5569-74, 2007 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-17376864

RESUMEN

Fifteen rare cancer-derived mutants of PIK3CA, the gene coding for the catalytic subunit p110alpha of phosphatidylinositol 3-kinase (PI3K), were examined for their biological and biochemical properties. Fourteen of these mutants show a gain of function: they induce rapamycin-sensitive oncogenic transformation of chicken embryo fibroblasts, constitutively activate Akt and TOR-mediated signaling, and show enhanced lipid kinase activity. Mapping of these mutants on a partial structural model of p110alpha suggests three groups of mutants, defined by their location in distinct functional domains of the protein. We hypothesize that each of these three groups induces a gain of PI3K function by a different molecular mechanism. Mutants in the C2 domain increase the positive surface charge of this domain and therefore may enhance the recruitment of p110alpha to cellular membranes. Mutants in the helical domain map to a contiguous surface of the protein and may affect the interaction with other protein(s). Mutants in the kinase domain are located near the hinge of the activation loop. They may alter the position and mobility of the activation loop. Arbitrarily introduced mutations that have no detectable phenotype map either to the interior of the protein or are positioned on a surface region that lies opposite to the exposed surfaces containing gain-of-function mutants. Engineered mutants that exchange acidic or neutral residues for basic residues on the critical surfaces show a gain of function.


Asunto(s)
Mutación , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Animales , Dominio Catalítico , Embrión de Pollo , Fosfatidilinositol 3-Quinasa Clase I , Activación Enzimática , Humanos , Lípidos/química , Modelos Moleculares , Fosfatidilinositol 3-Quinasas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
15.
J Neurosci ; 22(11): 4456-67, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12040053

RESUMEN

Calcium transients play an important role in the early and later phases of differentiation and maturation of single neurons and neuronal networks. Small-conductance calcium-activated potassium channels of the SK type modulate membrane excitability and are important determinants of the firing properties of central neurons. Increases in the intracellular calcium concentration activate SK channels, leading to a hyperpolarization of the membrane potential, which in turn reduces the calcium inflow into the cell. This feedback mechanism is ideally suited to regulate the spatiotemporal occurrence of calcium transients. However, the role of SK channels in neuronal development has not been addressed so far. We have concentrated on the ontogenesis and function of SK channels in the developing rat cerebellum, focusing particularly on Purkinje neurons. Electrophysiological recordings combined with specific pharmacological tools have revealed for the first time the presence of an afterhyperpolarizing current (I(AHP)) in immature Purkinje cells in rat cerebellar slices. The channel subunits underlying this current were identified as SK2 and localized by in situ hybridization and subunit-specific antibodies. Their expression level was shown to be high at birth and subsequently to decline during the first 3 weeks of postnatal life, both at the mRNA and protein levels. This developmental regulation was tightly correlated with the expression of I(AHP) and the prominent role of SK2 channels in shaping the spontaneous firing pattern in young, but not in adult, Purkinje neurons. These results provide the first evidence of the developmental regulation and function of SK channels in central neurons.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Canales de Potasio Calcio-Activados , Canales de Potasio/metabolismo , Subunidades de Proteína , Células de Purkinje/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Envejecimiento/metabolismo , Animales , Apamina/farmacología , Calcio/farmacología , Agonistas de los Canales de Calcio/farmacología , Señalización del Calcio/fisiología , Línea Celular , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Riñón/citología , Riñón/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/genética , Células de Purkinje/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Tetrodotoxina/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...