Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Physiol Cell Physiol ; 321(3): C549-C558, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34106787

RESUMEN

Cell signaling pathways regulating myosin regulatory light chain (LC20) phosphorylation contribute to determining contractile responses in smooth muscles. Following excitation and contraction, phasic smooth muscles, such as the digestive tract and urinary bladder, undergo relaxation due to a decline of cellular Ca2+ concentration and decreased Ca2+ sensitivity of LC20 phosphorylation, named Ca2+ desensitization. Here, we determined the mechanisms underlying the temporal Ca2+ desensitization of LC20 phosphorylation in phasic smooth muscles using permeabilized strips of the mouse ileum and urinary bladder. Upon stimulation with pCa6.0 at 20°C, contraction and LC20 phosphorylation peaked within 30 s and then declined to about 50% of the peak force at 2 min after stimulation. During the relaxation phase after the contraction, LC20 kinase [myosin light chain kinase (MLCK)] was inactivated, but no fluctuation in LC20 phosphatase activity occurred, suggesting that MLCK inactivation is a cause of the Ca2+-induced Ca2+ desensitization of LC20 phosphorylation. MLCK inactivation was associated with phosphorylation at the calmodulin-binding domain of the kinase. Treatment with STO-609 and TIM-063 antagonists for Ca2+/calmodulin (CaM)-dependent protein kinase kinase-ß (CaMKKß) attenuated both the phasic response of the contraction and MLCK phosphorylation, whereas neither CaM kinase II, AMP-activated protein kinase, nor p21-activated kinase induced MLCK inactivation in phasic smooth muscles. Conversely, protein phosphatase 2A inhibition amplified the phasic response. Signaling pathways through CaMKKß and protein phosphatase 2A may contribute to regulating the phasic response of smooth muscle contraction.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Músculo Liso Vascular/metabolismo , Cadenas Ligeras de Miosina/genética , Quinasa de Cadena Ligera de Miosina/genética , Proteína Fosfatasa 2/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Bencimidazoles/farmacología , Calcio/metabolismo , Señalización del Calcio , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Femenino , Regulación de la Expresión Génica , Íleon/metabolismo , Ratones , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso Vascular/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalimidas/farmacología , Fosforilación , Proteína Fosfatasa 2/metabolismo , Técnicas de Cultivo de Tejidos , Vejiga Urinaria/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
2.
J Cell Physiol ; 233(1): 434-446, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28295256

RESUMEN

Intercellular communication among autonomic nerves, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs) plays a central role in an uninterrupted regulation of blood flow through vascular contractile machinery. Impairment of this communication is linked to development of vascular diseases such as hypertension, cerebral/coronary vasospasms, aortic aneurism, and erectile dysfunction. Although the basic concept of the communication as a whole has been studied, the spatiotemporal correlation of ECs/VSMCs in tissues at the cellular level is unknown. Here, we show a unique VSMC response to ECs during contraction and relaxation of isolated aorta tissues through visualization of spatiotemporal activation patterns of smooth muscle myosin II. ECs in the intimal layer dictate the stimulus-specific heterogeneous activation pattern of myosin II in VSMCs within distinct medial layers. Myosin light chain (MLC) phosphorylation (active form of myosin II) gradually increases towards outer layers (approximately threefold higher MLC phosphorylation at the outermost layer than that of the innermost layer), presumably by release of an intercellular messenger, nitric oxide (NO). Our study also demonstrates that the MLC phosphorylation at the outermost layer in spontaneously hypertensive rats (SHR) during NO-induced relaxation is quite high and approximately 10-fold higher than that of its counterpart, the Wister-Kyoto rats (WKY), suggesting that the distinct pattern of myosin II activation within tissues is important for vascular protection against elevated blood pressure.


Asunto(s)
Células Endoteliales/fisiología , Técnica del Anticuerpo Fluorescente , Microscopía Fluorescente , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Vasoconstricción , Animales , Aorta Torácica/metabolismo , Aorta Torácica/fisiología , Biomarcadores , Comunicación Celular , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Técnicas In Vitro , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Miosina Tipo II/metabolismo , Óxido Nítrico/metabolismo , Fosforilación , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factores de Tiempo , Vasodilatación
3.
Pflugers Arch ; 469(12): 1651-1662, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28717991

RESUMEN

Prolonged bed rest (PBR) causes orthostatic hypotension (OH). Rapid constriction of splanchnic resistance arteries in response to a sudden increase in sympathetic tone contributes to the recovery of orthostatic arterial pressure upon standing. However, the molecular mechanism of PBR-induced dysfunction in arterial constriction is not fully understood. Previously, we showed that CPI-17, a regulatory protein for myosin phosphatase, mediates α1A-adrenergic receptor-induced rapid contraction of small mesenteric arteries. Here, we tested whether PBR associated with OH affects the α1-adrenergic receptor-induced CPI-17 signaling pathway in mesenteric arteries using rats treated by head-down tail-suspension hindlimb unloading (HDU), an experimental OH model. In normal anesthetized rats, mean arterial pressure (MAP) rapidly reduced upon 90° head-up tilt from supine position and then immediately recovered without change in heart rate, suggesting a rapid arterial constriction. On the other hand, after a 4-week HDU treatment, the fast orthostatic MAP recovery failed for 1 min. Alpha1A subtype-specific antagonist suppressed the orthostatic MAP recovery with a small decrease in basal blood pressure, whereas non-specific α1-antagonist prazosin strongly reduced both basal MAP and orthostatic recovery. The HDU treatment resulted in 68% reduction in contraction in parallel with 83% reduction in CPI-17 phosphorylation in denuded mesenteric arteries 10 s after α1-agonist stimulation. The treatment with either Ca2+-release channel opener or PKC inhibitor mimicked the deficiency in HDU arteries. These results suggest that an impairment of the rapid PKC/CPI-17 signaling pathway downstream of α1A-adrenoceptors in peripheral arterial constriction, as an end organ of orthostatic blood pressure reflex, is associated with OH in prolonged bed rest patients.


Asunto(s)
Reposo en Cama/efectos adversos , Hipotensión Ortostática/metabolismo , Arterias Mesentéricas/metabolismo , Proteínas Musculares/metabolismo , Fosfoproteínas/metabolismo , Animales , Presión Arterial/fisiología , Femenino , Inclinación de Cabeza/efectos adversos , Inclinación de Cabeza/fisiología , Frecuencia Cardíaca/fisiología , Hipotensión Ortostática/etiología , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley
4.
Pflugers Arch ; 465(2): 283-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23149489

RESUMEN

Smooth muscle caldesmon (h-CaD) is an actin- and myosin-binding protein that reversibly inhibits the actomyosin ATPase activity in vitro. To test the function of h-CaD in vivo, we eliminated its expression in mice. The h-CaD-null animals appeared normal and fertile, although the litter size was smaller. Tissues from the homozygotes lacked h-CaD and exhibited upregulation of the non-muscle isoform, l-CaD, in visceral, but not vascular tonic smooth muscles. While the Ca(2+) sensitivity of force generation of h-CaD-deficient smooth muscle remained largely unchanged, the kinetic behavior during relaxation in arteries was different. Both intact and permeabilized arterial smooth muscle tissues from the knockout animals relaxed more slowly than those of the wild type. Since this difference occurred after myosin dephosphorylation was complete, the kinetic effect most likely resulted from slower detachment of unphosphorylated crossbridges. Detailed analyses revealed that the apparently slower relaxation of h-CaD-null smooth muscle was due to an increase in the amplitude of a slower component of the biphasic tension decay. While the identity of this slower process has not been unequivocally determined, we propose it reflects a thin filament state that elicits fewer re-attached crossbridges. Our finding that h-CaD modulates the rate of smooth muscle relaxation clearly supports a role in the control of vascular tone.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Relajación Muscular/genética , Músculo Liso Vascular/metabolismo , Animales , Arterias/metabolismo , Arterias/patología , Arterias/fisiopatología , Proteínas de Unión a Calmodulina/metabolismo , Homocigoto , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miosinas/metabolismo , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación hacia Arriba , Vasodilatación/genética
5.
Prostate ; 73(9): 996-1006, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23389830

RESUMEN

BACKGROUND: Prostate smooth muscle plays an important role in the physiological ejection of prostatic fluid and also in the pathogenesis of benign prostate hyperplasia. Although mouse is the best genetically engineered animal model to identify potential molecular targets for human diseases, only fragmentary information is available for basic mechanism of mouse prostate contraction. METHODS: Small smooth muscle tubular rings were excised from four mouse prostate lobes to measure their isometric contractions. High K(+) , noradrenaline (NA), or acetylcholine (ACh) was applied with and without various antagonists and/or inhibitors to examine the contractile signaling pathways. RESULTS: Maximum amplitude of agonist-induced contractions varied greatly with different lobes but not with different locations or orientations within each lobe. Both NA and ACh produced large contractions in ventral and dorsal rings, whereas only small contractions were elicited in lateral and anterior rings. Combination of alpha-1 and muscarinic antagonists suppressed K(+) depolarization-induced contraction potently in ventral rings, but slightly in anterior rings. Blocking of either Ca(2+) -release or Ca(2+) -influx reduced agonist-induced contraction of ventral rings, however, a considerable amount of contractility remained even with both blockers. Inhibitors of ROCK and PKC partially inhibited NA-induced contractions, whereas a combination of Ca(2+) -blockers and Ca(2+) -sensitization inhibitors strongly suppressed the contraction. CONCLUSIONS: The ejection of prostatic fluid is differentially regulated in each prostate lobe. In ventral prostate smooth muscle, Ca(2+) -release, Ca(2+) -influx, and ROCK- and PKC-mediated Ca(2+) -sensitizations are all involved in NA-induced contractions. This finding is a useful step toward the understanding of the phenotypic changes in the smooth muscle of BPH prostate.


Asunto(s)
Músculo Liso/fisiología , Próstata/fisiología , Acetilcolina/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Atropina/farmacología , Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Agonistas Colinérgicos/farmacología , Técnicas In Vitro , Masculino , Ratones , Antagonistas Muscarínicos/farmacología , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso/anatomía & histología , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Nicardipino/farmacología , Norepinefrina/farmacología , Prazosina/farmacología , Próstata/anatomía & histología , Próstata/efectos de los fármacos , Próstata/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Rianodina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
6.
J Physiol ; 590(21): 5401-23, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22930267

RESUMEN

Each segment along arterial vessels adapts to different circumstances, including blood pressure and sympathetic innervation. PKC and Rho-associated kinase (ROCK) Ca(2+)-sensitizing pathways leading to myosin phosphatase inhibition are critically involved in α(1)-adrenoceptor-mediated vascular smooth muscle contraction in distinctive time-dependent manners. We tested whether the amplitude and time course of each pathway varies dynamically between arterial segments. Using pharmacological approaches, we determined the time-dependent roles of Ca(2+) release, Ca(2+) influx, PKC and ROCK in α(1)-agonist-induced contraction and phosphorylation of key proteins in denuded rat small mesenteric artery, midsized caudal artery and thoracic aorta. SR Ca(2+) release and voltage-dependent Ca(2+) influx were essential for the initial rising and late sustained phases, respectively, of phenylephrine-induced contraction, regardless of arterial size. In small mesenteric arteries, α(1A)-subtype-specific antagonists and inhibitors of PKC, but not ROCK, markedly reduced the initial and late phases of contraction in a non-additive manner and suppressed phosphorylation of myosin light chain (MLC) and CPI-17, but not myosin targeting subunit of myosin light chain phosphatase (MYPT1). In aorta, an α(1D)-specific antagonist reduced both the initial and late phases of contraction with a significant decrease in MLC but not CPI-17 or MYPT1 phosphorylation. ROCK inhibitors, but not PKC inhibitors, suppressed the sustained phase of contraction with a decrease in MLC and MYPT1 phosphorylation in the aorta. The effect of ROCK inhibitors was additive with the α(1D)-antagonist. The results for midsized arteries were intermediate. Thus, the PKC-CPI-17 Ca(2+)-sensitizing pathway, which is dependent on PKC subtype and a Ca(2+)-handling mechanism, and is downstream of α(1A) receptors, plays a major role in α(1)-agonist-induced contraction of small resistance arteries in the splanchnic vascular beds. The effect of PKC and ROCK increases and decreases, respectively, with decreasing arterial size.


Asunto(s)
Arterias/fisiología , Calcio/fisiología , Músculo Liso Vascular/fisiología , Proteína Quinasa C/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Quinasas Asociadas a rho/fisiología , Amidas/farmacología , Animales , Arterias/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Técnicas In Vitro , Masculino , Músculo Liso Vascular/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores
7.
Circ Res ; 107(10): 1220-31, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20884878

RESUMEN

RATIONALE: A homozygous disruption or genetic mutation of the bag3 gene, a member of the Bcl-2-associated athanogene (BAG) family proteins, causes cardiomyopathy and myofibrillar myopathy that is characterized by myofibril and Z-disc disruption. However, the detailed disease mechanism is not yet fully understood. OBJECTIVE: bag3(-/-) mice exhibit differences in the extent of muscle degeneration between muscle groups with muscles experiencing the most usage degenerating at an accelerated rate. Usage-dependent muscle degeneration suggests a role for BAG3 in supporting cytoskeletal connections between the Z-disc and myofibrils under mechanical stress. The mechanism by which myofibrillar structure is maintained under mechanical stress remains unclear. The purpose of the study is to clarify the detailed molecular mechanism of BAG3-mediated muscle maintenance under mechanical stress. METHODS AND RESULTS: To address the question of whether bag3 gene knockdown induces myofibrillar disorganization caused by mechanical stress, in vitro mechanical stretch experiments using rat neonatal cardiomyocytes and a short hairpin RNA-mediated gene knockdown system of the bag3 gene were performed. As expected, mechanical stretch rapidly disrupts myofibril structures in bag3 knockdown cardiomyocytes. BAG3 regulates the structural stability of F-actin through the actin capping protein, CapZß1, by promoting association between Hsc70 and CapZß1. BAG3 facilitates the distribution of CapZß1 to the proper location, and dysfunction of BAG3 induces CapZ ubiquitin-proteasome-mediated degradation. Inhibition of CapZß1 function by overexpressing CapZß2 increased myofibril vulnerability and fragmentation under mechanical stress. On the other hand, overexpression of CapZß1 inhibits myofibrillar disruption in bag3 knockdown cells under mechanical stress. As a result, heart muscle isolated from bag3(-/-) mice exhibited myofibrillar degeneration and lost contractile activity after caffeine contraction. CONCLUSIONS: These results suggest novel roles for BAG3 and Hsc70 in stabilizing myofibril structure and inhibiting myofibrillar degeneration in response to mechanical stress. These proteins are possible targets for further research to identify therapies for myofibrillar myopathy or other degenerative diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína CapZ/metabolismo , Proteínas Portadoras/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Mecanotransducción Celular , Miocardio/metabolismo , Miofibrillas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Cafeína/farmacología , Proteína CapZ/genética , Proteínas Portadoras/genética , Células Cultivadas , Humanos , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Contracción Miocárdica , Miocardio/patología , Miofibrillas/efectos de los fármacos , Miofibrillas/patología , Estabilidad Proteica , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Estrés Mecánico , Transfección
8.
J Smooth Muscle Res ; 58(0): 22-33, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35418530

RESUMEN

CPI-17 regulates the myosin phosphatase and mediates the agonist-induced contraction of smooth muscle. PKC and ROCK phosphorylate CPI-17 at Thr38 leading to a conformational change of the central inhibitory domain (PHIN domain). The N- and C-terminal tails of CPI-17 are predicted as unstructured loops and their sequences are conserved among mammals. Here we characterized CPI-17 N- and C-terminal unstructured tails using recombinant proteins that lack the potions. Recombinant CPI-17 proteins at a physiologic level (10 µM) were doped into beta-escin-permeabilized smooth muscle strips for Ca2+ sensitization force measurement. The ectopic full-length CPI-17 augmented the PDBu-induced Ca2+ sensitization force at pCa6.3, indicating myosin phosphatase inhibition. Deletion of N- and C-terminal tails of CPI-17 attenuated the extent of PDBu-induced Ca2+-sensitization force. The N-terminal deletion dampened phosphorylation at Thr38 by protein kinase C (PKC), and the C-terminal truncation lowered the affinity to the myosin phosphatase. Under the physiologic conditions, PKC and myosin phosphatase may recognize CPI-17 N-/C-terminal unstructured tails inducing Ca2+ sensitization force in smooth muscle cells.


Asunto(s)
Contracción Muscular , Proteínas Musculares , Animales , Calcio/metabolismo , Mamíferos/metabolismo , Contracción Muscular/fisiología , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo
9.
J Muscle Res Cell Motil ; 32(2): 65-76, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21643972

RESUMEN

Chronic treatment with fetal bovine serum (FBS) causes contractility reduction, morphological alteration and DNA synthesis in organ-cultured vascular tissues. Here, we tested the hypothesis that chronic inhibition of ROCK has a protective effect on FBS-induced alterations in small arteries. Rabbit mesenteric arterial rings were cultured in FBS-supplemented culture medium with or without Y-27632, a reversible ROCK inhibitor. Chronic Y-27632 treatment prevented FBS-induced gradual arterial constriction, wall thickening, reduced contractility, and increased ROCK-specific MYPT1 Thr853 phosphorylation. Treatment with Y-27632 also prevented decreased eNOS mRNA expression, and reduced acetylcholine-induced relaxation. Sudden application of Y-27632 to pre-cultured rings reduced MYPT1 phosphorylation and re-widened the constricted rings. Chronic treatment with Y-27632, however, rather augmented than reduced the FBS-induced RhoA over-expression, also increased ROCK1 and MYPT1 expression and averted the FBS-induced reduction of MLC expression, suggesting a compensation of inhibited RhoA/ROCK activity. Sudden removal of Y-27632 caused a rebound in MYPT1 phosphorylation and vasoconstriction in rabbit mesenteric artery. To test which ROCK isoform has greater involvement in FBS-induced contraction, haploinsufficient Rock1+/- and Rock2+/- mouse mesenteric arterial rings were subjected to organ-culture. FBS-induced contraction and RhoA over-expression in either heterozygous animal was not different from wild-type animals. These results suggest that FBS-induced contraction is mediated by up-regulation of RhoA and subsequent activation of ROCK. In conclusion, chronic ROCK inhibition produces some effects that protect against FBS-stimulated vasoconstriction and remodeling. There are also negative effects that a sudden withdrawal of ROCK inhibitor might cause a stronger vasoconstriction than before it was used.


Asunto(s)
Amidas/farmacología , Inhibidores Enzimáticos/farmacología , Arterias Mesentéricas/enzimología , Músculo Liso Vascular/enzimología , Piridinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Bovinos , Constricción Patológica/enzimología , Constricción Patológica/patología , Arterias Mesentéricas/patología , Ratones , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/patología , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Conejos , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
10.
Biochem Biophys Res Commun ; 401(1): 75-8, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20833141

RESUMEN

CPI-17 is a unique phosphoprotein that specifically inhibits myosin light chain phosphatase in smooth muscle and plays an essential role in agonist-induced contraction. To elucidate the in situ mechanism for G protein-mediated Ca²+-sensitization of CPI-17 phosphorylation, α-toxin-permeabilized arterial smooth muscle strips were used to monitor both force development and CPI-17 phosphorylation in response to GTPγS with varying Ca²+ concentrations. CPI-17 phosphorylation increased at unphysiologically high Ca²+ levels of pCa ≤ 6. GTPγS markedly enhanced the Ca²+ sensitivity of CPI-17 steady-state phosphorylation but had no enhancing effect under Ca²+-free conditions, while the potent PKC activator PDBu increased CPI-17 phosphorylation regardless of Ca²+ concentration. CPI-17 phosphorylation induced by pCa 4.5 alone was markedly inhibited by the presence of PKC inhibitor but not ROCK inhibitor. In the presence of calyculin A, a potent PP1/PP2A phosphatase inhibitor, CPI-17 phosphorylation increased with time even under Ca²+-free conditions. Furthermore, as Ca²+ concentration increased, so did CPI-17 phosphorylation rate. GTPγS markedly enhanced the rate of phosphorylation of CPI-17 at a given Ca²+. In the absence of calyculin A, either steady-state phosphorylation of CPI-17 under Ca²+-free conditions in the presence of GTPγS or at pCa 6.7 in the absence of GTPγS was negligible, suggesting a high intrinsic CPI-17 phosphatase activity. In conclusion, cooperative increases in Ca²+ and G protein activation are required for a significant activation of total kinases that phosphorylate CPI-17, which together overcome CPI-17 phosphatase activity and effectively increase the Ca²+ sensitivity of CPI-17 phosphorylation and smooth muscle contraction.


Asunto(s)
Calcio/metabolismo , Proteínas de Unión al GTP/metabolismo , Contracción Muscular , Proteínas Musculares/metabolismo , Músculo Liso Vascular/fisiología , Fosfoproteínas/metabolismo , Animales , Permeabilidad Capilar , Células Cultivadas , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Músculo Liso Vascular/enzimología , Fosforilación , Conejos
11.
J Physiol ; 587(Pt 14): 3587-603, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19470783

RESUMEN

Nitric oxide (NO) from endothelium is a major mediator of vasodilatation through cGMP/PKG signals that lead to a decrease in Ca(2+) concentration. In addition, NO-mediated signals trigger an increase in myosin light chain phosphatase (MLCP) activity. To evaluate the mechanism of NO-induced relaxation through MLCP deinhibition, we compared time-dependent changes in Ca(2+), myosin light chain (MLC) phosphorylation and contraction to changes in phosphorylation levels of CPI-17 at Thr38, RhoA at Ser188, and MYPT1 at Ser695, Thr696 and Thr853 in response to sodium nitroprusside (SNP)-induced relaxation in denuded rabbit femoral artery. During phenylephrine (PE)-induced contraction, SNP reduced CPI-17 phosphorylation to a minimal value within 15 s, in parallel with decreases in Ca(2+) and MLC phosphorylation, followed by a reduction of contractile force having a latency period of about 15 s. MYPT1 phosphorylation at Ser695, the PKG-target site, increased concurrently with relaxation. Phosphorylation of RhoA, MYPT1 Thr696 and Thr853 differed significantly at 5 min but not within 1 min of SNP exposure. Inhibition of Ca(2+) release delayed SNP-induced relaxation while inhibition of Ca(2+) channel, BK(Ca) channel or phosphodiesterase-5 did not. Pretreatment of resting artery with SNP suppressed an increase in Ca(2+), contractile force and phosphorylation of MLC, CPI-17, MYPT1 Thr696 and Thr853 at 10 s after PE stimulation, but had no effect on phorbol ester-induced CPI-17 phosphorylation. Together, these results suggest that NO production suppresses Ca(2+) release, which causes an inactivation of PKC and rapid CPI-17 dephosphorylation as well as MLCK inactivation, resulting in rapid MLC dephosphorylation and relaxation.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Óxido Nítrico/fisiología , Fosfoproteínas Fosfatasas/metabolismo , Animales , Calcio/metabolismo , Retroalimentación/fisiología , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Proteínas Musculares , Fosforilación , Conejos , Vasodilatación/fisiología
12.
Circ Res ; 100(1): 121-9, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17158339

RESUMEN

Ca(2+) ion is a universal intracellular messenger that regulates numerous biological functions. In smooth muscle, Ca(2+) with calmodulin activates myosin light chain (MLC) kinase to initiate a rapid MLC phosphorylation and contraction. To test the hypothesis that regulation of MLC phosphatase is involved in the rapid development of MLC phosphorylation and contraction during Ca(2+) transient, we compared Ca(2+) signal, MLC phosphorylation, and 2 modes of inhibition of MLC phosphatase, phosphorylation of CPI-17 Thr38 and MYPT1 Thr853, during alpha(1) agonist-induced contraction with/without various inhibitors in intact rabbit femoral artery. Phenylephrine rapidly induced CPI-17 phosphorylation from a negligible amount to a peak value of 0.38+/-0.04 mol of Pi/mol within 7 seconds following stimulation, similar to the rapid time course of Ca(2+) rise and MLC phosphorylation. This rapid CPI-17 phosphorylation was dramatically inhibited by either blocking Ca(2+) release from the sarcoplasmic reticulum or by pretreatment with protein kinase C inhibitors, suggesting an involvement of Ca(2+)-dependent protein kinase C. This was followed by a slow Ca(2+)-independent and Rho-kinase/protein kinase C-dependent phosphorylation of CPI-17. In contrast, MYPT1 phosphorylation had only a slow component that increased from 0.29+/-0.09 at rest to the peak of 0.68+/-0.14 mol of Pi/mol at 1 minute, similar to the time course of contraction. Thus, there are 2 components of the Ca(2+) sensitization through inhibition of MLC phosphatase. Our results support the hypothesis that the initial rapid Ca(2+) rise induces a rapid inhibition of MLC phosphatase coincident with the Ca(2+)-induced MLC kinase activation to synergistically initiate a rapid MLC phosphorylation and contraction in arteries with abundant CPI-17 content.


Asunto(s)
Calcio/metabolismo , Arteria Femoral/fisiología , Músculo Liso Vascular/fisiología , Vasoconstricción/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio , Arteria Femoral/efectos de los fármacos , Arteria Femoral/metabolismo , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Musculares/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fenilefrina/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Conejos , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Quinasas Asociadas a rho
13.
Structure ; 15(12): 1591-602, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18073109

RESUMEN

Phosphorylation of endogenous inhibitor proteins for type-1 Ser/Thr phosphatase (PP1) provides a mechanism for reciprocal coordination of kinase and phosphatase activities. A myosin phosphatase inhibitor protein CPI-17 is phosphorylated at Thr38 through G-protein-mediated signals, resulting in a >1000-fold increase in inhibitory potency. We show here the solution NMR structure of phospho-T38-CPI-17 with rmsd of 0.36 +/- 0.06 A for the backbone secondary structure, which reveals how phosphorylation triggers a conformational change and exposes an inhibitory surface. This active conformation is stabilized by the formation of a hydrophobic core of intercalated side chains, which is not formed in a phospho-mimetic D38 form of CPI-17. Thus, the profound increase in potency of CPI-17 arises from phosphorylation, conformational change, and hydrophobic stabilization of a rigid structure that poses the phosphorylated residue on the protein surface and restricts its hydrolysis by myosin phosphatase. Our results provide structural insights into transduction of kinase signals by PP1 inhibitor proteins.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Inhibidores Enzimáticos/química , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Fosforilación , Conformación Proteica , Homología de Secuencia de Aminoácido , Porcinos
14.
J Smooth Muscle Res ; 53(0): 1-19, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28260704

RESUMEN

A hallmark of smooth muscle cells is their ability to adapt their functions to meet temporal and chronic fluctuations in their demands. These functions include force development and growth. Understanding the mechanisms underlying the functional plasticity of smooth muscles, the major constituent of organ walls, is fundamental to elucidating pathophysiological rationales of failures of organ functions. Also, the knowledge is expected to facilitate devising innovative strategies that more precisely monitor and normalize organ functions by targeting individual smooth muscles. Evidence has established a current paradigm that the myosin light chain phosphatase (MLCP) is a master regulator of smooth muscle responsiveness to stimuli. Cellular MLCP activity is negatively and positively regulated in response to G-protein activation and cAMP/cGMP production, respectively, through the MYPT1 regulatory subunit and an endogenous inhibitor protein named CPI-17. In this article we review the outcomes from two decade of research on the CPI-17 signaling and discuss emerging paradoxes in the view of signaling pathways regulating smooth muscle functions through MLCP.


Asunto(s)
Contracción Muscular/genética , Músculo Liso/fisiología , Fosfatasa de Miosina de Cadena Ligera/fisiología , Transducción de Señal/fisiología , Calcio/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Musculares , Tono Muscular/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas Fosfatasas/fisiología , Fosforilación , Transducción de Señal/genética , Quinasas Asociadas a rho/fisiología
15.
Int J Clin Exp Pathol ; 5(8): 787-95, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071861

RESUMEN

During the influenza pandemic of 2009, the number of viral pneumonia cases showed a marked increase in comparison with seasonal influenza viruses. Mutations at amino acid 222 (D222G mutations) in the virus hemagglutinin (HA) molecule, known to alter the receptor-recognition properties of the virus, were detected in a number of the more severely-affected patients in the early phases of the pandemic. To understand the background for the emergence of the mutant amino acid D222G in human lungs, we conducted histological examinations on lung specimens of patients from Mexico who had succumbed in the pandemic. Prominent regenerative and hyperplastic changes in the alveolar type II pneumocytes, which express avian-type sialoglycan receptors in the respiratory tract of severely affected individuals, were observed in the Mexican patients. An infection model utilizing guinea pigs, which was chosen in order to best simulate the sialic acid distribution of severe pneumonia in human patients, demonstrated an increase of D222G mutants and a delay in the diminution of mutants in the lower respiratory tract in comparison to the upper respiratory tract. Our data suggests that the predominance of avian-type sialoglycan receptors in the pneumonic lungs may contribute to the emergence of viral HA mutants. This data comprehensively illustrates the mechanisms for the emergence of mutants in the clinical samples.


Asunto(s)
Modelos Animales de Enfermedad , Hemaglutininas Virales/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Neumonía Viral/virología , Células Epiteliales Alveolares/virología , Animales , Brotes de Enfermedades , Femenino , Genes Virales , Cobayas , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/patología , Células de Riñón Canino Madin Darby , Mutación , Neumonía Viral/patología , ARN Viral/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
PLoS One ; 6(6): e21207, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21701593

RESUMEN

Avian influenza H5N1 and pandemic (H1N1) 2009 viruses are known to induce viral pneumonia and subsequent acute respiratory distress syndrome (ARDS) with diffuse alveolar damage (DAD). The mortality rate of ARDS/DAD is extremely high, at approximately 60%, and no effective treatment for ARDS/DAD has been established. We examined serial pathological changes in the lungs of mice infected with influenza virus to determine the progress from viral pneumonia to ARDS/DAD. Mice were intranasally infected with influenza A/Puerto Rico/8/34 (PR8) virus, and their lungs were examined both macro- and micro-pathologically every 2 days. We also evaluated general condition, survival rate, body weight, viral loads in lung, and surfactant proteins in serum. As a result, all infected mice died within 9 days postinfection. At 2 days postinfection, inflammation in alveolar septa, i.e., interstitial pneumonia, was observed around bronchioles. From 4 to 6 days postinfection, interstitial pneumonia with alveolar collapse expanded throughout the lungs. From 6 to 9 days postinfection, DAD with severe alveolar collapse was observed in the lungs of all of dying and dead mice. In contrast, DAD was not observed in the live infected-mice from 2 to 6 days postinfection, despite their poor general condition. In addition, histopathological analysis was performed in mice infected with a dose of PR8 virus which was 50% of the lethal dose for mice in the 20-day observation period. DAD with alveolar collapse was observed in all dead mice. However, in the surviving mice, instead of DAD, glandular metaplasia was broadly observed in their lungs. The present study indicates that DAD with severe alveolar collapse is associated with death in this mouse infection model of influenza virus. Inhibition of the development of DAD with alveolar collapse may decrease the mortality rate in severe viral pneumonia caused by influenza virus infection.


Asunto(s)
Virus de la Influenza A/patogenicidad , Pulmón/patología , Pulmón/virología , Infecciones por Orthomyxoviridae/patología , Animales , Línea Celular , Perros , Femenino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología
17.
Development ; 134(18): 3327-38, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17720696

RESUMEN

Heparan sulfate (HS) plays an essential role in extracellular signaling during development. Biochemical studies have established that HS binding to ligands and receptors is regulated by the fine 6-O-sulfated structure of HS; however, mechanisms that control sulfated HS structure and associated signaling functions in vivo are not known. Extracellular HS 6-O-endosulfatases, SULF1 and SULF2, are candidate enzymatic regulators of HS 6-O-sulfated structure and modulate HS-dependent signaling. To investigate Sulf regulation of developmental signaling, we have disrupted Sulf genes in mouse and identified redundant functions of Sulfs in GDNF-dependent neural innervation and enteric glial formation in the esophagus, resulting in esophageal contractile malfunction in Sulf1(-/-);Sulf2(-/-) mice. SULF1 is expressed in GDNF-expressing esophageal muscle and SULF2 in innervating neurons, establishing their direct functions in esophageal innervation. Biochemical and cell signaling studies show that Sulfs are the major regulators of HS 6-O-desulfation, acting to reduce GDNF binding to HS and to enhance GDNF signaling and neurite sprouting in the embryonic esophagus. The functional specificity of Sulfs in GDNF signaling during esophageal innervation was established by showing that the neurite sprouting is selectively dependent on GDNF, but not on neurotrophins or other signaling ligands. These findings provide the first in vivo evidence that Sulfs are essential developmental regulators of cellular HS 6-O-sulfation for matrix transmission and reception of GDNF signal from muscle to innervating neurons.


Asunto(s)
Esófago/embriología , Esófago/inervación , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Sistema Nervioso Periférico/embriología , Sulfatasas/fisiología , Sulfotransferasas/fisiología , Animales , Esófago/metabolismo , Heparitina Sulfato/metabolismo , Ratones , Ratones Mutantes , Contracción Muscular , Músculo Esquelético/fisiología , Miocitos del Músculo Liso/fisiología , Neuritas/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Transducción de Señal , Sulfatasas/genética , Sulfotransferasas/genética
18.
J Cell Sci ; 119(Pt 9): 1769-80, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16608882

RESUMEN

Phosphorylation of myosin light chain (MLC) and contraction of differentiated smooth muscle cells in vascular walls are regulated by Ca2+ -dependent activation of MLC kinase, and by Rho-kinase- or protein-kinases-C-dependent inhibition of MLC phosphatase (MLCP). We examined regulatory pathways for MLC kinase and MLCP in cultured vascular smooth muscle cells (VSMCs), and for isometric force generation of VSMCs reconstituted in collagen fibers. Protein levels of RhoA, Rho-kinase and MYPT1 (a regulatory subunit of MLCP) were upregulated in cultured VSMCs, whereas a MLCP inhibitor protein, CPI-17, was downregulated. Endothelin-1 evoked a steady rise in levels of Ca2+, MLC phosphorylation and the contractile force of VSMCs, whereas angiotensin-II induced transient signals. Also, Thr853 phosphorylation of MYPT1 occurred in response to stimuli, but neither agonist induced phosphorylation of MYPT1 at Thr696. Unlike fresh aortic tissues, removal of Ca2+ or addition of voltage-dependent Ca2+ -channel blocker did not inhibit contractions of reconstituted VSMC fibers induced by agonists or even high concentrations of extracellular K+ ions. Inhibitors of Ins(1,4,5)P3-receptor and Rho-kinase antagonized agonist-induced or high-K+ -induced contraction in both reconstituted fibers and fresh tissues. These results indicate that both Ins(1,4,5)P3-induced Ca2+ release and Rho-kinase-induced MYPT1 phosphorylation at Thr853 play pivotal roles in MLC phosphorylation of cultured VSMCs where either Ca2+ -influx or CPI-17-MLCP signaling is downregulated.


Asunto(s)
Contracción Isométrica/fisiología , Potenciales de la Membrana/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Transducción de Señal/fisiología , Angiotensina II/metabolismo , Compuestos de Anilina/metabolismo , Animales , Aorta/citología , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Células Cultivadas , Endotelina-1/metabolismo , Colorantes Fluorescentes/metabolismo , Masculino , Miocitos del Músculo Liso/citología , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fenilefrina/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Proteína Fosfatasa 1 , Ratas , Ratas Sprague-Dawley , Vasoconstrictores/metabolismo , Xantenos/metabolismo
19.
J Physiol ; 546(Pt 3): 879-89, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12563012

RESUMEN

Myosin phosphatase (MLCP) plays a critical regulatory role in the Ca(2+) sensitivity of myosin phosphorylation and smooth muscle contraction. It has been suggested that phosphorylation at Thr(695) of the MLCP regulatory subunit (MYPT1) and at Thr(38) of the MLCP inhibitor protein CPI-17 results in inhibition of MLCP activity. We have previously demonstrated that CPI-17 Thr(38) phosphorylation plays an important role in G-protein-mediated inhibition of MLCP in tonic arterial smooth muscle. Here, we attempted to evaluate the function of MYPT1 in phasic rabbit portal vein (PV) and vas deferens (VD) smooth muscles. Using site- and phospho-specific antibodies, phosphorylation of MYPT1 Thr(695) and CPI-17 Thr(38) was examined along with MYPT1 Thr(850), which is a non-inhibitory Rho-kinase site. We found that both CPI-17 Thr(38) and MYPT1 Thr(850) were phosphorylated in response to agonists or GTPgammaS concurrently with contraction and myosin phosphorylation in alpha-toxin-permeabilized PV tissues. In contrast, phosphorylation of MYPT1 Thr(695) did not increase. Comparable results were also obtained in both permeabilized and intact VD. The Rho-kinase inhibitor Y-27632 and the protein kinase C (PKC) inhibitor GF109203X suppressed phosphorylation of MYPT1 Thr(850) and CPI-17 Thr(38), respectively, in intact VD while MYPT1 Thr(695) phosphorylation was insensitive to both inhibitors. These results indicate that phosphorylation of MYPT1 Thr(695) is independent of stimulation of G-proteins, Rho-kinase or PKC. In the phasic PV, phosphorylation of CPI-17 Thr(38) may contribute towards inhibition of MLCP while the phasic visceral VD, which has a low CPI-17 concentration, probably utilizes other Ca(2+) sensitizing mechanisms for inhibiting MLCP besides phosphorylation of MYPT1 and CPI-17.


Asunto(s)
Calcio/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas/metabolismo , Amidas/farmacología , Animales , Electrofisiología , Endotelina-1/farmacología , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Isoenzimas/metabolismo , Masculino , Maleimidas/farmacología , Músculo Liso Vascular/fisiología , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera , Permeabilidad , Fenilefrina/farmacología , Fosforilación , Vena Porta/fisiología , Piridinas/farmacología , Conejos , Fosfolipasas de Tipo C/farmacología , Conducto Deferente/fisiología , Vasoconstricción
20.
J Physiol ; 557(Pt 2): 515-28, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15090608

RESUMEN

Ca(2+) sensitivity of arterial contractility is governed by regulating myosin phosphatase activity in response to agonist stimuli. CPI-17, a myosin phosphatase inhibitor phosphoprotein, is phosphorylated concomitantly with agonist-induced contractile Ca(2+) sensitization in mammalian artery. CPI-17 has not been detected in chicken artery, but is readily detectable in pigeon artery. To evaluate a role of CPI-17, we compared contractility of the arteries of 'CPI-17-deficient' chicken with those of CPI-17-rich rabbit and pigeon, and studied the effect of CPI-17-reconstitution in chicken artery. Other major regulatory/contractile proteins for Ca(2+) sensitization are expressed in both chicken and rabbit arteries. Agonists, such as an alpha(1)-agonist and endothelin-1, produced significant contraction in arteries of all species under physiological Ca(2+)-containing conditions. Depletion of Ca(2+) abolished these contractions in chicken but partially inhibited them in rabbit and pigeon arteries. Unlike CPI-17-rich tissues, chicken arteries exerted little Ca(2+) sensitization in response to alpha(1)-agonist or endothelin-1. GTPgammaS produced a slight Ca(2+) sensitizing effect in chicken artery, but this was significantly smaller compared with CPI-17-rich tissues. A PKC activator (PDBu) did not generate but rather reduced a contraction in both intact and alpha-toxin-permeabilized chicken artery in contrast to a large contraction in CPI-17-rich arteries. Myosin light chain phosphorylation was reduced by PDBu in chicken but elevated in rabbit artery. Addition of recombinant CPI-17 into beta-escin-permeabilized chicken artery restored PDBu-induced and enhanced GTPgammaS-induced Ca(2+) sensitization. Thus, CPI-17 is essential for G protein/PKC-mediated Ca(2+) sensitization in smooth muscle.


Asunto(s)
Calcio/metabolismo , Pollos/fisiología , Proteínas de Unión al GTP/metabolismo , Proteínas Musculares/deficiencia , Músculo Liso/fisiología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfoproteínas/deficiencia , Proteína Quinasa C/metabolismo , Animales , Aorta , Pollos/metabolismo , Columbidae , Masculino , Contracción Muscular/fisiología , Proteínas Musculares/biosíntesis , Proteínas Musculares/inmunología , Músculo Liso/enzimología , Cadenas Ligeras de Miosina/metabolismo , Fosfoproteínas/biosíntesis , Fosfoproteínas/inmunología , Fosforilación , Conejos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA