Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nutrients ; 15(7)2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37049598

RESUMEN

Intestinal epithelial barrier function is closely associated with the development of many intestinal diseases. Heat-killed Lacticaseibacillus paracasei (HK-LP) has been shown to improve intestinal health and enhance immunity. However, the function of HK-LP in the intestinal barrier is still unclear. This study characterized the inflammatory effects of seven HK-LP (1 µg/mL) on the intestinal barrier using lipopolysaccharide (LPS) (100 µg/mL)-induced Caco-2 cells. In this study, HK-LP 6105, 6115, and 6235 were selected, and their effects on the modulation of inflammatory factors and tight junction protein expression (claudin-1, zona occludens-1, and occludin) were compared. The effect of different cultivation times (18 and 48 h) was investigated in response to LPS-induced intestinal epithelial barrier dysfunction. Our results showed that HK-LP 6105, 6115, and 6235 improved LPS-induced intestinal barrier permeability reduction and transepithelial resistance. Furthermore, HK-LP 6105, 6115, and 6235 inhibited the pro-inflammatory factors (TNF-α, IL-1ß, IL-6) and increased the expression of the anti-inflammatory factors (IL-4, IL-10, and TGF-ß). HK-LP 6105, 6115, and 6235 ameliorated the inflammatory response. It inhibited the nuclear factor kappa B (NF-κB) signaling pathway-mediated myosin light chain (MLC)/MLC kinase signaling pathway by downregulating the Toll-like receptor 4 (TLR4)/NF-κB pathway. Thus, the results suggest that HK-LP 6150, 6115, and 6235 may improve intestinal health by regulating inflammation and TJ proteins. Postbiotics produced by these strains exhibit anti-inflammatory properties that can protect the intestinal barrier.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Humanos , FN-kappa B/metabolismo , Células CACO-2 , Lipopolisacáridos/farmacología , Cadenas Ligeras de Miosina , Lacticaseibacillus , Mucosa Intestinal/metabolismo , Calor , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Fosforilación , Antiinflamatorios/farmacología , Uniones Estrechas/metabolismo
2.
Ecotoxicol Environ Saf ; 257: 114940, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37099960

RESUMEN

Fluoride is a common contaminant of groundwater and agricultural commodity, which poses challenges to animal and human health. A wealth of research has demonstrated its detrimental effects on intestinal mucosal integrity; however, the underlying mechanisms remain obscure. This study aimed to investigate the role of the cytoskeleton in fluoride-induced barrier dysfunction. After sodium fluoride (NaF) treatment of the cultured Caco-2 cells, both cytotoxicity and cytomorphological changes (internal vacuoles or massive ablation) were observed. NaF lowered transepithelial electrical resistance (TEER) and enhanced paracellular permeation of fluorescein isothiocyanate dextran 4 (FD-4), indicating Caco-2 monolayers hyperpermeability. In the meantime, NaF treatment altered both the expression and distribution of the tight junction protein ZO-1. Fluoride exposure increased myosin light chain II (MLC2) phosphorylation and triggered actin filament (F-actin) remodeling. While inhibition of myosin II by Blebbistatin blocked NaF-induced barrier failure and ZO-1 discontinuity, the corresponding agonist Ionomycin had effects comparable to those of fluoride, suggesting that MLC2 serves as an effector. Given the mechanisms upstream of p-MLC2 regulation, further studies demonstrated that NaF activated RhoA/ROCK signaling pathway and myosin light chain kinase (MLCK), strikingly increasing the expression of both. Pharmacological inhibitors (Rhosin, Y-27632 and ML-7) reversed NaF-induced barrier breakdown and stress fiber formation. The role of intracellular calcium ions ([Ca2+]i) in NaF effects on Rho/ROCK pathway and MLCK was investigated. We found that NaF elevated [Ca2+]i, whereas chelator BAPTA-AM attenuated increased RhoA and MLCK expression as well as ZO-1 rupture, thus, restoring barrier function. Collectively, abovementioned results suggest that NaF induces barrier impairment via Ca2+-dependent RhoA/ROCK pathway and MLCK, which in turn triggers MLC2 phosphorylation and rearrangement of ZO-1 and F-actin. These results provide potential therapeutic targets for fluoride-induced intestinal injury.


Asunto(s)
Fluoruros , Quinasa de Cadena Ligera de Miosina , Animales , Humanos , Fosforilación , Células CACO-2 , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Fluoruros/metabolismo , Calcio/metabolismo , Actinas/metabolismo , Uniones Estrechas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
3.
Aquat Toxicol ; 257: 106424, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36863152

RESUMEN

Aquafeeds are susceptible to contamination caused by aflatoxin B1 (AFB1). The gill of fish is an important respiratory organ. However, few studies have investigated the effects of dietary AFB1 exposure on gill. This study aimed to discuss the effects of AFB1 on the structural and immune barrier of grass carp gill. Dietary AFB1 increased reactive oxygen species (ROS) levels, protein carbonyl (PC) and malondialdehyde (MDA) contents, which consequently caused oxidative damage. In contrast, dietary AFB1 decreased antioxidant enzymes activities, relative genes expression (except MnSOD) and the contents of glutathione (GSH) (P < 0.05), which are partly regulated by NF-E2-related factor 2 (Nrf2/Keap1a). Moreover, dietary AFB1 caused DNA fragmentation. The relative genes of apoptosis (except Bcl-2, McL-1 and IAP) were significantly upregulated (P < 0.05), and apoptosis was likely upregulated through p38 mitogen-activated protein kinase (p38MAPK). The relative expressions of genes associated with tight junction complexes (TJs) (except ZO-1 and claudin-12) were significantly decreased (P < 0.05), and TJs were likely regulated by myosin light chain kinase (MLCK). Overall, dietary AFB1 disrupted the structural barrier of gill. Furthermore, AFB1 increased gill sensitivity to F. columnare, increased Columnaris disease and decreased the production of antimicrobial substances (P < 0.05) in grass carp gill, and upregulated the expression of genes involved with pro-inflammatory factors (except TNF-α and IL-8) and the pro-inflammatory response partly attributed to the regulation by nuclear factor κB (NF-κB). Meanwhile, the anti-inflammatory factors were downregulated (P < 0.05) in grass carp gill after challenge with F. columnare, which was partly attributed to the target of rapamycin (TOR). These results suggested that AFB1 aggravated the disruption of the immune barrier of grass carp gill after being challenge with F. columnare. Finally, the upper limit of safety of AFB1 for grass carp, based on Columnaris disease, was 31.10 µg/kg diet.


Asunto(s)
Carpas , Contaminantes Químicos del Agua , Animales , FN-kappa B/metabolismo , Suplementos Dietéticos/análisis , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Aflatoxina B1/toxicidad , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Carpas/metabolismo , Branquias/metabolismo , Inmunidad Innata , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Contaminantes Químicos del Agua/toxicidad , Transducción de Señal , Dieta/veterinaria , Antioxidantes/metabolismo , Glutatión , Alimentación Animal/análisis
4.
Int J Mol Sci ; 24(3)2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36768366

RESUMEN

Mechanical properties of healthy and Dupuytren fibroblasts were investigated by atomic force microscopy (AFM). In addition to standard force curves, rheological properties were assessed using an oscillatory testing methodology, in which the frequency was swept from 1 Hz to 1 kHz, and data were analyzed using the structural damping model. Dupuytren fibroblasts showed larger apparent Young's modulus values than healthy ones, which is in agreement with previous results. Moreover, cell mechanics were compared before and after ML-7 treatment, which is a myosin light chain kinase inhibitor (MLCK) that reduces myosin activity and hence cell contraction. We employed two different concentrations of ML-7 inhibitor and could observe distinct cell reactions. At 1 µM, healthy and scar fibroblasts did not show measurable changes in stiffness, but Dupuytren fibroblasts displayed a softening and recovery after some time. When increasing ML-7 concentration (3 µM), the majority of cells reacted, Dupuytren fibroblasts were the most susceptible, not being able to recover from the drug and dying. These results suggested that ML-7 is a potent inhibitor for MLCK and that myosin II is essential for cytoskeleton stabilization and cell survival.


Asunto(s)
Citoesqueleto , Contractura de Dupuytren , Fibroblastos , Microscopía de Fuerza Atómica , Contracción Muscular , Cadenas Ligeras de Miosina , Humanos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Contractura de Dupuytren/tratamiento farmacológico , Contractura de Dupuytren/metabolismo , Contractura de Dupuytren/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fenómenos Mecánicos , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Quinasa de Cadena Ligera de Miosina/uso terapéutico , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología
5.
Gene ; 851: 147030, 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36351549

RESUMEN

Advanced glycation end products (AGEs) disturb endothelial barrier function and contribute to age-related diseases. As microRNAs (miRNAs) are potential therapeutic agents, targeting AGEs-associated signaling using miRNAs in endothelial cells may be an effective intervention strategy for age-related vascular disorders. This study investigated the effects of AGEs on the endothelial cell senescence and barrier function in human umbilical vein endothelial cells (HUVECs). HUVECs were treated with AGEs and transfected with miRNA-1-3p mimics to induce overexpression of miR-1-3p. Senescence-associated ß-galactosidase (SA-ß-Gal) staining and senescence-related proteins P53, P21, and P16 were detected to evaluate the endothelial cell senescence. The expression levels of myosin light chain kinase (MLCK) signaling and transendothelial electric resistance (TEER) were used to indicate endothelial barrier function. AGEs significantly increased SA-ß-gal staining-positive cells accompanied by the upregulation of P53, P21, and P16 expression. AGEs also damaged endothelial barrier function by decreasing TEER and increasing zonula occludens protein 1, p-MLC/MLC, and MLCK. miRNA-1-3p was significantly reduced in HUVECs treated with AGEs. miR-1-3p overexpression decreased MLCK signal and improved AGEs-induced endothelial barrier function impairment. Meanwhile, miR-1-3p overexpression ameliorated oxidative stress and endothelial cell senescence induced by AGEs. AGEs induced endothelial cell senescence and endothelial barrier dysfunction by regulating miR-1-3p/MLCK signaling pathway.


Asunto(s)
MicroARNs , Quinasa de Cadena Ligera de Miosina , Humanos , Senescencia Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Productos Finales de Glicación Avanzada/metabolismo
6.
Psychopharmacology (Berl) ; 236(5): 1583-1596, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31147734

RESUMEN

RATIONALE: Intestinal permeability plays an important role in gut-brain axis communication. Recent studies indicate that intestinal permeability increases in neonate pups during maternal separation (MS). OBJECTIVES: The present study aims to determine whether pharmacological inhibition of myosin light chain kinase (MLCK), which regulates tight junction contraction and controls intestinal permeability, in stressed neonates, protects against the long-term effects of MS. METHODS: Male Wistar rats were exposed to MS (3 h per day from post-natal day (PND)2 to PND14) or left undisturbed and received daily intraperitoneal injection of a MLCK inhibitor (ML-7, 5 mg/kg) or vehicle during the same period. At adulthood, emotional behaviors, corticosterone response to stress, and gut microbiota composition were analyzed. RESULTS: ML-7 restored gut barrier function in MS rats specifically during the neonatal period. Remarkably, ML-7 prevented MS-induced sexual reward-seeking impairment and reversed the alteration of corticosterone response to stress at adulthood. The effects of ML-7 were accompanied by the normalization of the abundance of members of Lachnospiraceae, Clostridiales, Desulfovibrio, Bacteroidales, Enterorhabdus, and Bifidobacterium in the feces of MS rats at adulthood. CONCLUSIONS: Altogether, our work suggests that improvement of intestinal barrier defects during development may alleviate some of the long-term effects of early-life stress and provides new insight on brain-gut axis communication in a context of stress.


Asunto(s)
Azepinas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Privación Materna , Naftalenos/farmacología , Estrés Psicológico/metabolismo , Animales , Animales Recién Nacidos , Azepinas/uso terapéutico , Corticosterona/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Microbioma Gastrointestinal/fisiología , Masculino , Quinasa de Cadena Ligera de Miosina/farmacología , Quinasa de Cadena Ligera de Miosina/uso terapéutico , Naftalenos/uso terapéutico , Embarazo , Ratas , Ratas Wistar , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/psicología , Factores de Tiempo
7.
Biochem J ; 429(2): 291-302, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20459395

RESUMEN

KRP (kinase-related protein), also known as telokin, has been proposed to inhibit smooth muscle contractility by inhibiting the phosphorylation of the rMLC (regulatory myosin light chain) by the Ca2+-activated MLCK (myosin light chain kinase). Using the phosphatase inhibitor microcystin, we show in the present study that KRP also inhibits Ca2+-independent rMLC phosphorylation and smooth muscle contraction mediated by novel Ca2+-independent rMLC kinases. Incubating KRP-depleted Triton-skinned taenia coli with microcystin at pCa>8 induced a slow contraction reaching 90% of maximal force (Fmax) at pCa 4.5 after approximately 25 min. Loading the fibres with KRP significantly slowed down the force development, i.e. the time to reach 50% of Fmax was increased from 8 min to 35 min. KRP similarly inhibited rMLC phosphorylation of HMM (heavy meromyosin) in vitro by MLCK or by the constitutively active MLCK fragment (61K-MLCK) lacking the myosin-docking KRP domain. A C-terminally truncated KRP defective in myosin binding inhibited neither force nor HMM phosphorylation. Phosphorylated KRP inhibited the rMLC phosphorylation of HMM in vitro and Ca2+-insensitive contractions in fibres similar to unphosphorylated KRP, whereby the phosphorylation state of KRP was not altered in the fibres. We conclude that (i) KRP inhibits not only MLCK-induced contractions, but also those elicited by Ca2+-independent rMLC kinases; (ii) phosphorylation of KRP does not modulate this effect; (iii) binding of KRP to myosin is essential for this inhibition; and (iv) KRP inhibition of rMLC phosphorylation is most probably due to the shielding of the phosphorylation site on the rMLC.


Asunto(s)
Colon/fisiología , Contracción Muscular/fisiología , Quinasa de Cadena Ligera de Miosina/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Calcio/metabolismo , Calcio/farmacología , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/farmacología , Pollos , Colon/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cartilla de ADN/genética , Femenino , Cobayas , Humanos , Técnicas In Vitro , Masculino , Toxinas Marinas , Microcistinas/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/farmacología , Octoxinol , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
8.
J Physiol ; 588(Pt 6): 981-93, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20123786

RESUMEN

Phosphorylation of myosin regulatory light chain (RLC) by myosin light chain kinase (MLCK) and myosin binding protein-C (cMyBP-C) by protein kinase A (PKA) independently accelerate the kinetics of force development in ventricular myocardium. However, while MLCK treatment has been shown to increase the Ca(2+) sensitivity of force (pCa(50)), PKA treatment has been shown to decrease pCa(50), presumably due to cardiac troponin I phosphorylation. Further, MLCK treatment increases Ca(2+)-independent force and maximum Ca(2+)-activated force, whereas PKA treatment has no effect on either force. To investigate the structural basis underlying the kinase-specific differential effects on steady-state force, we used synchrotron low-angle X-ray diffraction to compare equatorial intensity ratios (I(1,1)/I(1,0)) to assess the proximity of myosin cross-bridge mass relative to actin and to compare lattice spacings (d(1,0)) to assess the inter-thick filament spacing in skinned myocardium following treatment with either MLCK or PKA. As we showed previously, PKA phosphorylation of cMyBP-C increases I(1,1)/I(1,0) and, as hypothesized, treatment with MLCK also increased I(1,1)/I(1,0), which can explain the accelerated rates of force development during activation. Importantly, interfilament spacing was reduced by 2 nm (3.5%) with MLCK treatment, but did not change with PKA treatment. Thus, RLC or cMyBP-C phosphorylation increases the proximity of cross-bridges to actin, but only RLC phosphorylation affects lattice spacing, which suggests that RLC and cMyBP-C modulate the kinetics of force development by similar structural mechanisms; however, the effect of RLC phosphorylation to increase the Ca(2+) sensitivity of force is mediated by a distinct mechanism, most probably involving changes in interfilament spacing.


Asunto(s)
Proteínas Portadoras/fisiología , Contracción Miocárdica/fisiología , Cadenas Ligeras de Miosina/fisiología , Función Ventricular Derecha/fisiología , Animales , Calcio/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos , Modelos Animales , Contracción Miocárdica/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Función Ventricular Derecha/efectos de los fármacos
9.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G361-70, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19541925

RESUMEN

As a regulator of smooth muscle contraction, zipper-interacting protein kinase (ZIPK) can directly phosphorylate the myosin regulatory light chains (LC20) and produce contractile force. Synthetic peptides (SM-1 and AV25) derived from the autoinhibitory region of smooth muscle myosin light chain kinase can inhibit ZIPK activity in vitro. Paradoxically, treatment of Triton-skinned ileal smooth muscle strips with AV25, but not SM-1, potentiated Ca2+-independent, microcystin- and ZIPK-induced contractions. The AV25-induced potentiation was limited to ileal and colonic smooth muscles and was not observed in rat caudal artery. Thus the potentiation of Ca2+-independent contractions by AV25 appeared to be mediated by a mechanism unique to intestinal smooth muscle. AV25 treatment elicited increased phosphorylation of LC20 (both Ser-19 and Thr-18) and myosin phosphatase-targeting subunit (MYPT1, inhibitory Thr-697 site), suggesting involvement of a Ca2+-independent LC20 kinase with coincident inhibition of myosin phosphatase. The phosphorylation of the inhibitor of myosin phosphatase, CPI-17, was not affected. The AV25-induced potentiation was abolished by pretreatment with staurosporine, a broad-specificity kinase inhibitor, but specific inhibitors of Rho-associated kinase, PKC, and MAPK pathways had no effect. When a dominant-negative ZIPK [kinase-dead ZIPK((1-320))-D161A] was added to skinned ileal smooth muscle, the potentiation of microcystin-induced contraction by AV25 was blocked. Furthermore, pretreatment of skinned ileal muscle with SM-1 abolished AV25-induced potentiation. We conclude, therefore, that, even though AV25 is an in vitro inhibitor of ZIPK, activation of the ZIPK pathway occurs following application of AV25 to permeabilized ileal smooth muscle. Finally, we propose a mechanism whereby conformational changes in the pseudosubstrate region of ZIPK permit augmentation of ZIPK activity toward LC(20) and MYPT1 in situ. AV25 or molecules based on its structure could be used in therapeutic situations to induce contractility in diseases of the gastrointestinal tract associated with hypomotility.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Motilidad Gastrointestinal/efectos de los fármacos , Íleon/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso/enzimología , Quinasa de Cadena Ligera de Miosina/metabolismo , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/química , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Dominio Catalítico , Pollos , Proteínas Quinasas Asociadas a Muerte Celular , Activación Enzimática , Íleon/enzimología , Técnicas In Vitro , Microcistinas/farmacología , Proteínas Musculares/metabolismo , Músculo Liso/efectos de los fármacos , Mutación , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteína Fosfatasa 1/metabolismo , Ratas , Especificidad por Sustrato
10.
J Mol Biol ; 390(5): 879-92, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19477187

RESUMEN

A current popular model to explain phosphorylation of smooth muscle myosin (SMM) by myosin light-chain kinase (MLCK) proposes that MLCK is bound tightly to actin but weakly to SMM. We found that MLCK and calmodulin (CaM) co-purify with unphosphorylated SMM from chicken gizzard, suggesting that they are tightly bound. Although the MLCK:SMM molar ratio in SMM preparations was well below stoichiometric (1:73+/-9), the ratio was approximately 23-37% of that in gizzard tissue. Fifteen to 30% of MLCK was associated with CaM at approximately 1 nM free [Ca(2+)]. There were two MLCK pools that bound unphosphorylated SMM with K(d) approximately 10 and 0.2 microM and phosphorylated SMM with K(d) approximately 20 and 0.2 microM. Using an in vitro motility assay to measure actin sliding velocities, we showed that the co-purifying MLCK-CaM was activated by Ca(2+) and phosphorylation of SMM occurred at a pCa(50) of 6.1 and at a Hill coefficient of 0.9. Similar properties were observed from reconstituted MLCK-CaM-SMM. Using motility assays, co-sedimentation assays, and on-coverslip enzyme-linked immunosorbent assays to quantify proteins on the motility assay coverslip, we provide strong evidence that most of the MLCK is bound directly to SMM through the telokin domain and some may also be bound to both SMM and to co-purifying actin through the N-terminal actin-binding domain. These results suggest that this MLCK may play a role in the initiation of contraction.


Asunto(s)
Calmodulina/metabolismo , Complejos Multiproteicos/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Miosinas del Músculo Liso/metabolismo , Actinas/metabolismo , Adenosina Trifosfato/farmacología , Animales , Bovinos , Pollos , Ensayo de Inmunoadsorción Enzimática , Humanos , Cinética , Magnesio/farmacología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/farmacología , Fragmentos de Péptidos/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína
11.
J Biol Chem ; 281(13): 8332-8, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16410249

RESUMEN

Calmodulin (CaM) is a ubiquitous Ca2+ sensor protein that plays an important role in regulating a large number of Ca2+ channels, including the inositol 1,4,5-trisphosphate receptor (IP3R). Despite many efforts, the exact mechanism by which CaM regulates the IP3R still remains elusive. Here we show, using unidirectional 45Ca2+ flux experiments on permeabilized L15 fibroblasts and COS-1 cells, that endogenously bound CaM is essential for the proper activation of the IP3R. Removing endogenously bound CaM by titration with a high affinity (pM) CaM-binding peptide derived from smooth muscle myosin light-chain kinase (MLCK peptide) strongly inhibited IP3-induced Ca2+ release. This inhibition was concentration- and time-dependent. Removing endogenously bound CaM affected the maximum release capacity but not its sensitivity to IP3. A mutant peptide with a strongly reduced affinity for CaM did not affect inhibited IP3-induced Ca2+ release. Furthermore, the inhibition by the MLCK peptide was fully reversible. Re-adding exogenous CaM, but not CaM1234, reactivated the IP3R. These data suggest that, by using a specific CaM-binding peptide, we removed endogenously bound CaM from a high affinity CaM-binding site on the IP3R, and this resulted in a complete loss of the IP3R activity. Our data support a new model whereby CaM is constitutively associated with the IP3R and functions as an essential subunit for proper functioning of the IP3R.


Asunto(s)
Canales de Calcio/metabolismo , Calmodulina/metabolismo , Calmodulina/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Sitios de Unión , Células COS , Calcio/análisis , Calcio/antagonistas & inhibidores , Calcio/metabolismo , Canales de Calcio/genética , Línea Celular , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato , Cinética , Pulmón/citología , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/farmacología , Oocitos/efectos de los fármacos , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Erizos de Mar/citología , Spodoptera/citología , Spodoptera/genética
12.
Am J Physiol Heart Circ Physiol ; 287(6): H2712-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15331360

RESUMEN

It is generally recognized that ventricular myosin regulatory light chains (RLC) are approximately 40% phosphorylated under basal conditions, and there is little change in RLC phosphorylation with agonist stimulation of myocardium or altered stimulation frequency. To establish the functional consequences of basal RLC phosphorylation in the heart, we measured mechanical properties of rat skinned trabeculae in which approximately 7% or approximately 58% of total RLC was phosphorylated. The protocol for achieving approximately 7% phosphorylation of RLC involved isolating trabeculae in the presence of 2,3-butanedione monoxime (BDM) to dephosphorylate RLC from its baseline level. Subsequent phosphorylation to approximately 58% of total was achieved by incubating BDM-treated trabeculae in solution containing smooth muscle myosin light chain kinase, calmodulin, and Ca2+ (i.e., MLCK treatment). After MLCK treatment, Ca2+ sensitivity of force increased by 0.06 pCa units and maximum force increased by 5%. The rate constant of force development (ktr) increased as a function of Ca2+ concentration in the range between pCa 5.8 and pCa 4.5. When expressed versus pCa, the activation dependence of ktr appeared to be unaffected by MLCK treatment; however, when activation was expressed in terms of isometric force-generating capability (as a fraction of maximum), MLCK treatment slowed ktr at submaximal activations. These results suggest that basal phosphorylation of RLC plays a role in setting the kinetics of force development and Ca2+ sensitivity of force in cardiac muscle. Our results also argue that changes in RLC phosphorylation in the range examined here influence actin-myosin interaction kinetics differently in heart muscle than was previously reported for skeletal muscle.


Asunto(s)
Calcio/metabolismo , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Animales , Femenino , Técnicas In Vitro , Cinética , Quinasa de Cadena Ligera de Miosina/farmacología , Fosforilación , Ratas , Ratas Wistar
13.
J Muscle Res Cell Motil ; 25(8): 657-65, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15750850

RESUMEN

Telokin, a 17 kDa smooth muscle specific protein, consists of the C-terminal domain of MLCK, is phosphorylated by PKA and PKG at Ser13 in vivo (Wu et al. (1998) J Biol Chem 273: 11362-11369; Walker et al. (2001) J. Biol Chem 276: 24519-24524) and is proposed to induce Ca2+-desensitization through activation of myosin phosphatase (Wu et al. (1998) J. Biol Chem 273: 11362-11369). Telokin is reported to be highly expressed in phasic with only trace amounts in tonic smooth muscle. In alpha-toxin permeabilized femoral artery, 5 microM 8-Br-cGMP induced a two-fold increase in telokin phosphorylation and a maximal 30% relaxation of Ca2+-activated force compared to a 90% relaxation in phasic ileum muscle consistent with the relative amounts of telokin expressed in ileum, 27+/-4.6 microM SEM compared to 6+/-1.7 microM SEM, in femoral artery. Recombinant Wt telokin and the phospho-telokin mutant, S13D relaxed telokin-depleted femoral artery, by 38+/-8% SEM and 60+/-20% SEM, respectively. 8-Br-cGMP increased the rate and decreased the amplitude of force development initiated by photolysis of caged ATP in alpha-toxin permeabilized ileum and femoral artery smooth muscle, consistent with a cGMP-induced increase in phosphatase activity. Similarly, in telokin depleted ileum, recombinant S13D mutant telokin significantly increased the rate (0.08+/-0.01 s-1 vs. 014+/-0.02 s-1) and decreased force amplitude. In conclusion, our data support a role for telokin in cyclic nucleotide-induced relaxation of not only phasic, but also tonic smooth muscle and that this relaxation is mediated by activation of myosin phosphatase activity leading to a decrease in myosin light chain phosphorylation.


Asunto(s)
Calcio/fisiología , Músculo Liso/fisiología , Quinasa de Cadena Ligera de Miosina/fisiología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Péptidos/fisiología , Adenosina Trifosfato/metabolismo , Animales , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Activación Enzimática , Masculino , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso/efectos de los fármacos , Músculo Liso/enzimología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Quinasa de Cadena Ligera de Miosina/biosíntesis , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/farmacología , Fragmentos de Péptidos , Péptidos/metabolismo , Péptidos/farmacología , Fosforilación , Conejos , Proteínas Recombinantes/farmacología , Factores de Tiempo
14.
Blood ; 99(6): 2060-9, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11877280

RESUMEN

Combretastatin A-4-phosphate (CA-4-P) is a tubulin-binding compound currently in clinical trial as a tumor vascular-targeting agent. In endothelial cells, CA-4-P is known to cause microtubule depolymerization, but little is known about its subsequent effects on cell morphology and function. Here, we demonstrate that within minutes of endothelial cell exposure to CA-4-P, myosin light chain (MLC) was phosphorylated, leading to actinomyosin contractility, assembly of actin stress fibers, and formation of focal adhesions. These cytoskeletal alterations appeared to be a consequence of Rho activation, as they were abolished by either the Rho inhibitor C3 exoenzyme or Rho-kinase inhibitor Y-27632. In response to CA-4-P, some cells rapidly assumed a blebbing morphology in which F-actin accumulated around surface blebs, stress fibers misassembled into a spherical network surrounding the cytoplasm, and focal adhesions appeared malformed. Blebbing was associated with decreased cell viability and could be inhibited by Rho/Rho-kinase inhibitors or by blocking the CA-4-P-mediated activation of stress-activated protein kinase-2/p38. The extracellular-regulated kinases 1 and 2 (ERK-1/2) were shown to protect against blebbing since blebbing was attenuated on ERK-1/2 stimulation and was up-regulated by specific inhibition of ERK-1/2 activation. The use of MLC kinase (MLCK) and myosin adenosine triphosphatase inhibitors led us to propose a role for MLCK and myosin activity independent of MLC phosphorylation in regulating the blebbing process. CA-4-P-mediated contractility and blebbing were associated with a Rho-dependent increase in monolayer permeability to dextrans, suggesting that such functional changes may be important in the rapid response of the tumor endothelium to CA-4-P in vivo.


Asunto(s)
Actinas/efectos de los fármacos , Antineoplásicos Fitogénicos/farmacología , Citoesqueleto/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/ultraestructura , Estilbenos/farmacología , Actinas/metabolismo , Proteínas de Fase Aguda/efectos de los fármacos , Proteínas de Fase Aguda/farmacología , Proteínas de Fase Aguda/fisiología , Permeabilidad Capilar/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Endotelio Vascular/citología , Adhesiones Focales/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/farmacología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Quinasa de Cadena Ligera de Miosina/farmacología , Quinasa de Cadena Ligera de Miosina/fisiología , Miosinas/farmacología , Miosinas/fisiología , Necrosis , Fibras de Estrés/efectos de los fármacos
15.
J Neurosci ; 21(21): 8464-72, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11606635

RESUMEN

The postsynaptic density (PSD) at excitatory dendritic synapses comprises a protein complex of glutamate receptors, scaffolding elements, and signaling enzymes. For example, NMDA receptors (NMDARs) are linked to several proteins in the PSD, such as PSD-95, and are also tethered via binding proteins such as alpha-actinin directly to filamentous actin of the cytoskeleton. Depolymerization of the cytoskeleton modulates the activity of NMDARs, and, in turn, strong activation of NMDARs can trigger depolymerization of actin. Myosin, the motor protein of muscular contraction and nonmuscle motility, is also associated with NMDARs and the PSD. We show here that constitutively active myosin light chain kinase (MLCK) enhances NMDAR-mediated whole-cell and synaptic currents in acutely isolated CA1 pyramidal and cultured hippocampal neurons, whereas inhibitors of MLCK depress these currents. This MLCK-dependent regulation was observed in cell-attached patches but was lost after excision to inside-out patches. Furthermore, the enhancement induced by constitutively active MLCK and the depression of MLCK inhibitors were eliminated after depolymerization of the cytoskeleton. NMDARs and MLCK did not colocalize in clusters on the dendrites of cultured hippocampal neurons, further indicating that the effects of MLCK are mediated indirectly via actomyosin. Our results suggest that MLCK enhances actomyosin contractility to either increase the membrane tension on NMDARs or to alter physical relationships between the actin cytoskeleton and the linker proteins of NMDARs.


Asunto(s)
Actinas/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Calcio/metabolismo , Carbocianinas , Separación Celular , Células Cultivadas , Dendritas/metabolismo , Inhibidores Enzimáticos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Colorantes Fluorescentes , Hipocampo , Ratones , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/farmacología , N-Metilaspartato/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Células Piramidales/citología , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/efectos de los fármacos
16.
J Biol Chem ; 276(43): 39797-804, 2001 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-11514555

RESUMEN

To understand how the plasma membrane Ca(2+) pump (PMCA) behaves under changing Ca(2+) concentrations, it is necessary to obtain information about the Ca(2+) dependence of the rate constants for calmodulin activation (k(act)) and for inactivation by calmodulin removal (k(inact)). Here we studied these constants for isoforms 2b and 4b. We measured the ATPase activity of these isoforms expressed in Sf9 cells. For both PMCA4b and 2b, k(act) increased with Ca(2+) along a sigmoidal curve. At all Ca(2+) concentrations, 2b showed a faster reaction with calmodulin than 4b but a slower off rate. On the basis of the measured rate constants, we simulated mathematically the behavior of these pumps upon repetitive changes in Ca(2+) concentration and also tested these simulations experimentally; PMCA was activated by 500 nm Ca(2+) and then exposed to 50 nm Ca(2+) for 10 to 150 s, and then Ca(2+) was increased again to 500 nm. During the second exposure to 500 nm Ca(2+), the activity reached steady state faster than during the first exposure at 500 nm Ca(2+). This memory effect is longer for PMCA2b than for 4b. In a separate experiment, a calmodulin-binding peptide from myosin light chain kinase, which has no direct interaction with the pump, was added during the second exposure to 500 nm Ca(2+). The peptide inhibited the activity of PMCA2b when the exposure to 50 nm Ca(2+) was 150 s but had little or no effect when this exposure was only 15 s. This suggests that the memory effect is due to calmodulin remaining bound to the enzyme during the period at low Ca(2+). The memory effect observed in PMCA2b and 4b will allow cells expressing either of them to remove Ca(2+) more quickly in subsequent spikes after an initial activating spike.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Calcio/metabolismo , Membrana Celular/enzimología , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/genética , Calmodulina/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Proteínas de Transporte de Catión , Humanos , Cinética , Modelos Teóricos , Quinasa de Cadena Ligera de Miosina/farmacología , Fragmentos de Péptidos/farmacología , ATPasas Transportadoras de Calcio de la Membrana Plasmática , Isoformas de Proteínas , Proteínas Recombinantes/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 281(2): G467-78, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11447027

RESUMEN

In smooth muscle cells enzymatically isolated from circular muscle of the esophagus (ESO) and lower esophageal sphincter (LES), ACh-induced contraction and myosin light chain (MLC) phosphorylation were similar. Contraction and phosphorylation induced by purified MLC kinase (MLCK) were significantly greater in LES than ESO. ACh-induced contraction and MLC phosphorylation were inhibited by calmodulin and MLCK inhibitors in LES and by protein kinase C (PKC) inhibitors in ESO. Contraction of LES and ESO induced by the PKC agonist 1,2-dioctanoylglycerol (DG) was unaffected by MLCK inhibitors. Caldesmon and calponin concentration-dependently inhibited ACh-induced contraction of ESO and not LES. In ESO, caldesmon antagonist GS17C reversed caldesmon- but not calponin-induced ACh inhibition. GS17C caused contraction of permeabilized ESO but had much less effect on LES. GS17C-induced contraction was not affected by MLCK inhibitors, suggesting that MLCK may not regulate caldesmon-mediated contraction. DG-induced contraction of ESO and LES was inhibited by caldesmon and calponinin, suggesting that these proteins may regulate PKC-dependent contraction. We conclude that calmodulin and MLCK play a role in ACh-induced LES contraction, whereas the classical MLCK may not be the major kinase responsible for contraction and phosphorylation of MLC in ESO. ESO contraction is PKC dependent. Caldesmon and/or calponin may play a role in PKC-dependent contraction.


Asunto(s)
Unión Esofagogástrica/fisiología , Esófago/fisiología , Contracción Muscular , Músculo Liso/fisiología , Quinasa de Cadena Ligera de Miosina/farmacología , Proteína Quinasa C/fisiología , Acetilcolina/farmacología , Animales , Proteínas de Unión al Calcio/farmacología , Calmodulina/fisiología , Proteínas de Unión a Calmodulina/farmacología , Gatos , Células Cultivadas , Femenino , Masculino , Proteínas de Microfilamentos , Músculo Liso/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación , Transducción de Señal , Calponinas
18.
J Cell Physiol ; 174(3): 370-9, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9462699

RESUMEN

Although the signaling pathways leading to hydrogen peroxide (H2O2)-induced endothelial monolayer permeability remain ambiguous, cytoskeletal proteins are known to be essential for maintaining endothelial integrity and regulating solute flux through the monolayer. We have recently demonstrated that thrombin-induced actin reorganization in bovine pulmonary artery endothelial cells (BPAEC) requires activation of both myosin light chain kinase (MLCK) and protein kinase C (PKC). Therefore, the present study was designed to investigate the effects of H2O2 on actin reorganization in BPAEC. H2O2 initiated sustained recruitment of actin to the cytoskeleton and transient myosin recruitment in a time- and concentration-dependent manner. The H2O2-induced actin recruitment was significantly inhibited by the calmodulin antagonists, W7 and TFP, but not by the MLCK inhibitor, KT5926, nor the PKC inhibitors, H7 and calphostin C. H2O2 also caused actin filament rearrangement in BPAEC with disruption of the dense peripheral bands and formation of stress fibers. These alterations occurred prior to actin translocation to the cytoskeleton and are prevented by inhibition of either MLCK or PKC. High concentrations of H2O2 transiently attenuated PKC activity but slightly increased the phosphorylation of the prominent PKC substrate and actin-binding protein, myristoylated alanine-rich C kinase substrate (MARCKS), by 5 min. However, MARCKS phosphorylation was reduced to below basal levels by 30 min. On the other hand, H2O2 induced a time- and dose-dependent phosphorylation of myosin light chains which was eliminated by both MLCK and PKC inhibitors. These data suggest that MLCK contributes to H2O2-induced myosin light chain phosphorylation and actin rearrangement and that PKC may play a permissive role. Neither of these enzymes appears to be involved in the H2O2-induced recruitment of actin to the cytoskeleton.


Asunto(s)
Citoesqueleto/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Actinas/metabolismo , Actinas/fisiología , Animales , Bovinos , Línea Celular , Citoesqueleto/enzimología , Citoesqueleto/fisiología , Endotelio Vascular/enzimología , Quinasa de Cadena Ligera de Miosina/farmacología , Miosinas/metabolismo , Miosinas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Arteria Pulmonar
19.
Neurosci Lett ; 197(1): 75-7, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8545061

RESUMEN

Actions of myosin light chain kinase inhibitors were tested on delayed rectifier potassium current (IK) in dissociated bullfrog sympathetic neurons. A microbial product, wortmannin (10 microM, extracellularly) and a synthetic peptide, SM-1 (20 microM, intracellularly) caused approximately 35 mV hyperpolarizing shift of the inactivation curve. Substitution of ATP (1.15 mM) in the pipette solution with 5'-adenylylimidodiphosphate mimicked the actions of wortmannin and SM-1. Results suggest that phosphorylation of myosin may modulate kinetics for the inactivation of IK.


Asunto(s)
Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Neuronas/metabolismo , Canales de Potasio/metabolismo , Sistema Nervioso Simpático/metabolismo , Adenilil Imidodifosfato/farmacología , Androstadienos/farmacología , Animales , Electrofisiología , Inhibidores Enzimáticos/farmacología , Ganglios Simpáticos/citología , Ganglios Simpáticos/efectos de los fármacos , Técnicas In Vitro , Quinasa de Cadena Ligera de Miosina/farmacología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Canales de Potasio/efectos de los fármacos , Rana catesbeiana , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/efectos de los fármacos , Wortmanina
20.
J Neurosci ; 14(12): 7695-703, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7996204

RESUMEN

Ca(2+)-induced exocytosis in chromaffin cells now seems to consist of at least two distinct steps:MgATP-dependent Ca(2+)-dependent priming of the secretory apparatus, and Ca(2+)-dependent MgATP-independent step that triggers exocytosis (Bittner and Holz, 1992). Recently we found that a specific inhibitor of myosin light chain kinase (MLCK), wortmannin, inhibits Ca(2+)-induced catecholamine release from digitonin-permeabilized chromaffin cells, suggesting an implication of MLCK in the mechanisms of Ca(2+)-induced exocytosis (Imaizumi et al., 1992b). To elucidate further the implication of MLCK in the mechanism of exocytosis, we studied the effects of wortmannin and a peptide inhibitor (SM-1) corresponding to the pseudosubstrate domain of MLCK on MgATP-dependent and MgATP-independent release in digitonin-permeabilized chromaffin cells. Ca(2+)-induced exocytosis from the permeabilized cells in the presence of MgATP was inhibited by both SM-1 and wortmannin. Inhibitory effect of wortmannin on the rate of release induced by 10 microM Ca2+ in the presence of MgATP was much prominent in the later phase (1-10 min), although the initial rate was also decreased. SM-1 strongly inhibited ATP-dependent release without affecting Ca(2+)-dependent ATP-independent release at all. In addition, priming effect of MgATP that underlies Ca(2+)-dependent ATP-independent release was remarkably reduced by both wortmannin and SM-1. These results suggest that MLCK plays an essential role in ATP-dependent priming of Ca(2+)-induced exocytosis in chromaffin cells.


Asunto(s)
Adenosina Trifosfato/fisiología , Glándulas Suprarrenales/fisiología , Sistema Cromafín/fisiología , Exocitosis , Quinasa de Cadena Ligera de Miosina/fisiología , Adenosina Trifosfato/farmacología , Glándulas Suprarrenales/citología , Androstadienos/farmacología , Animales , Calcio/metabolismo , Catecolaminas/metabolismo , Bovinos , Células Cultivadas , Sistema Cromafín/citología , Técnica del Anticuerpo Fluorescente , Immunoblotting , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/farmacología , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Factores de Tiempo , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA