Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Arch Immunol Ther Exp (Warsz) ; 70(1): 4, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35043239

RESUMEN

The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK's versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.


Asunto(s)
Neoplasias , Investigación con Células Madre , Animales , Desarrollo Embrionario , Ratones , Isoformas de Proteínas , Transducción de Señal , Quinasas Asociadas a rho/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(38): 18815-18821, 2019 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-31467166

RESUMEN

Flexible and low-cost poly(ethylene oxide) (PEO)-based electrolytes are promising for all-solid-state Li-metal batteries because of their compatibility with a metallic lithium anode. However, the low room-temperature Li-ion conductivity of PEO solid electrolytes and severe lithium-dendrite growth limit their application in high-energy Li-metal batteries. Here we prepared a PEO/perovskite Li3/8Sr7/16Ta3/4Zr1/4O3 composite electrolyte with a Li-ion conductivity of 5.4 × 10-5 and 3.5 × 10-4 S cm-1 at 25 and 45 °C, respectively; the strong interaction between the F- of TFSI- (bis-trifluoromethanesulfonimide) and the surface Ta5+ of the perovskite improves the Li-ion transport at the PEO/perovskite interface. A symmetric Li/composite electrolyte/Li cell shows an excellent cyclability at a high current density up to 0.6 mA cm-2 A solid electrolyte interphase layer formed in situ between the metallic lithium anode and the composite electrolyte suppresses lithium-dendrite formation and growth. All-solid-state Li|LiFePO4 and high-voltage Li|LiNi0.8Mn0.1Co0.1O2 batteries with the composite electrolyte have an impressive performance with high Coulombic efficiencies, small overpotentials, and good cycling stability.

3.
Nat Commun ; 9(1): 1809, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29728558

RESUMEN

Developing cost-effective electrocatalysts operated in the same electrolyte for water splitting, including oxygen and hydrogen evolution reactions, is important for clean energy technology and devices. Defects in electrocatalysts strongly influence their chemical properties and electronic structures, and can dramatically improve electrocatalytic performance. However, the development of defect-activated electrocatalyst with an efficient and stable water electrolysis activity in alkaline medium remains a challenge, and the understanding of catalytic origin is still limited. Here, we highlight defect-enriched bifunctional eletrocatalyst, namely, three-dimensional iron fluoride-oxide nanoporous films, fabricated by anodization/fluorination process. The heterogeneous films with high electrical conductivity possess embedded disorder phases in crystalline lattices, and contain numerous scattered defects, including interphase boundaries, stacking faults, oxygen vacancies, and dislocations on the surfaces/interface. The heterocatalysts efficiently catalyze water splitting in basic electrolyte with remarkable stability. Experimental studies and first-principle calculations suggest that the surface/edge defects contribute significantly to their high performance.

4.
Oncotarget ; 9(16): 12995-13008, 2018 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-29560126

RESUMEN

Doxorubicin is among the essential medicines with a wide antitumor spectrum, but its clinical application is limited by its cardiotoxicity. We recently discovered that ROCK1 is a key molecule in mediating cardiac remodeling in response to various stresses. To determine the roles of ROCK1 in doxorubicin cardiotoxicity, we gave three doses of doxorubicin injections to wild type (WT) and ROCK1-/- mice with one week intervals between treatments, the cumulative dose being 24 mg/kg. ROCK1-/- mice exhibited preserved cardiac function, reduced apoptosis, autophagy and fibrosis compared to the WT mice. To further determine the cellular mechanisms, we have examined the role of ROCK1 in cardiomyocytes using cardiomyocyte-specific knockout mice, MHC-Cre/ROCK1fl/fl, which partially reproduced the cardioprotective characteristics of ROCK1-/- mice, indicating that ROCK1 in both cardiomyocytes and non-cardiomyocytes mediates doxorubicin cardiotoxicity. To elucidate the molecular mechanisms, a detailed time course study after a single doxorubicin injection at 10 mg/kg was performed in ROCK1-/- and MHC-Cre/ROCK1fl/fl mice. The molecular analysis revealed that both ROCK1-/- and MHC-Cre/ROCK1fl/fl hearts exhibited significant reduction of doxorubicin-induced early responses including increased apoptotic (Bax) and autophagic (p62/SQSTM1 and LC3-II) markers, associated with reduced Beclin 1 phosphorylation on Thr119, supporting reduced Beclin 1-mediated autophagy initiation due to increased association of Beclin 1 with Bcl 2 or Bcl-XL in these hearts compared to the WT or ROCK1fl/fl mice. These results support that ROCK1 deficiency is cardioprotective against doxorubicin-induced cardiotoxicity at least in part through reducing Beclin 1-mediated autophagy initiation in cardiomyocytes and restoring autophagic flux to ameliorate doxorubicin cardiotoxicity.

5.
Arch Immunol Ther Exp (Warsz) ; 64(4): 259-78, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26725045

RESUMEN

Rho-associated coiled-coil kinase (ROCK) is a major downstream effector of the small GTPase RhoA. The ROCK family, consisting of ROCK1 and ROCK2, plays a central role in the organization of the actin cytoskeleton, and is involved in a wide range of fundamental cellular functions such as contraction, adhesion, migration, proliferation, and apoptosis. Since the discovery of effective inhibitors such as fasudil and Y27632, the biological roles of ROCK have been extensively explored in numerous diseases, including cancer. Accumulating evidence supports the concept that ROCK plays important roles in tumor development and progression through regulating many key cellular functions associated with malignancy, including tumorigenicity, tumor growth, metastasis, angiogenesis, tumor cell apoptosis/survival and chemoresistance as well. This review focuses on the new advances of the most recent 5 years from the studies on the roles of ROCK in cancer development and progression; the discussion is mainly focused on the potential value of ROCK inhibitors in cancer therapy.


Asunto(s)
Neoplasias/metabolismo , Quinasas Asociadas a rho/metabolismo , Apoptosis , Adhesión Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , MicroARNs/metabolismo , Mutación , Metástasis de la Neoplasia , Neoplasias/genética , Células Madre Neoplásicas/citología , Neovascularización Patológica , Polimorfismo Genético , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/genética
6.
Hypertension ; 67(3): 597-605, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26781283

RESUMEN

The insufficiency of compensatory angiogenesis in the heart of patients with hypertension contributes to heart failure transition. The hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling cascade controls responsive angiogenesis. One of the challenges in reprograming the insufficient angiogenesis is to achieve a sustainable tissue exposure to the proangiogenic factors, such as HIF1α stabilization. In this study, we identified Rnd3, a small Rho GTPase, as a proangiogenic factor participating in the regulation of the HIF1α-VEGF signaling cascade. Rnd3 physically interacted with and stabilized HIF1α, and consequently promoted VEGFA expression and endothelial cell tube formation. To demonstrate this proangiogenic role of Rnd3 in vivo, we generated Rnd3 knockout mice. Rnd3 haploinsufficient (Rnd3(+/-)) mice were viable, yet developed dilated cardiomyopathy with heart failure after transverse aortic constriction stress. The poststress Rnd3(+/-) hearts showed significantly impaired angiogenesis and decreased HIF1α and VEGFA expression. The angiogenesis defect and heart failure phenotype were partially rescued by cobalt chloride treatment, a HIF1α stabilizer, confirming a critical role of Rnd3 in stress-responsive angiogenesis. Furthermore, we generated Rnd3 transgenic mice and demonstrated that Rnd3 overexpression in heart had a cardioprotective effect through reserved cardiac function and preserved responsive angiogenesis after pressure overload. Finally, we assessed the expression levels of Rnd3 in the human heart and detected significant downregulation of Rnd3 in patients with end-stage heart failure. We concluded that Rnd3 acted as a novel proangiogenic factor involved in cardiac responsive angiogenesis through HIF1α-VEGFA signaling promotion. Rnd3 downregulation observed in patients with heart failure may explain the insufficient compensatory angiogenesis involved in the transition to heart failure.


Asunto(s)
Vasos Coronarios/patología , Regulación de la Expresión Génica , Hipertensión/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Unión al GTP rho/genética , Animales , Western Blotting , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Neovascularización Patológica/metabolismo , ARN/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis
7.
J BUON ; 20(5): 1223-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26537068

RESUMEN

PURPOSE: Prostate cancer (PCa) is one of the most common malignancies in males, and multiple genetic studies have confirmed association with susceptibility to PCa. However, the risk conferred in men living in China is unkown. We selected 6 previously identified variants as candidates to define their association with PCa in Chinese men. METHODS: We genotyped 6 single nucleotide polymorphisms (SNPs) (rs1465618, rs1983891, rs339331, rs16901966, rs1447295 and rs10090154) using high resolution melting (HRM) analysis and assessed their association with PCa risk in a case-control study of 481 patients and 480 controls in a Chinese population. In addition, the individual and cumulative contribution for the risk of PCa and clinical covariates were analysed. RESULTS: We found that 5 of the 6 genetic variants were associated with PCa risk. The T allele of rs339331 and the G allele of rs16901966 showed a significant association with PCa susceptibility: OR (95%CI)= 0.78 (0.64-0.94), p<0.009 and OR (95%CI)= 0.66 (0.54-0.81), p<0.0001, as well as A allele of rs1447295 (OR [95%CI]=1.46 (1.17-1.84), p<0.001) and T allele of rs10090154 (OR [95%CI]= 0.58 (0.46-0.74), p<0.0001). rs339331(T) was associated with a 0.71-fold and 1.42-fold increase of PCa risk by dominant model (p=0.007) and recessive model (p=0.007). rs16901966 (G) was associated with a 0.51-fold and 1.98-fold increase of PCa risk by dominant model (p=0.006) and recessive model (p=0.0058). rs10090154 (T) was associated with a 1.89-fold and 0.53-fold increase of PCa risk by dominant model (p=0.000006) and recessive model (p=0.000006). And, rs1983891(C) was associated with a 0.77-fold increase of PCa risk by recessive model (p=0.045). rs1447295 was associated with a 1.57-fold increase of PCa risk by dominant model (p=0.008). rs1465618 showed no significant association with PCa. The cumulative effects test of risk alleles (rs rs1983891, rs339331, rs16901966, rs1447295 and rs10090154) showed an increasing risk to PCa in a frequency-dependent manner (ptrend=0.001), and men with more than 3 risk alleles had the most significant susceptibility to PCa (OR=1.99, p=0.001), compared with those who had one risk allele (OR=1.17, p=0.486). CONCLUSION: Our results provide further support for association of the THADA, FOXP4, GPRC6A/RFX6 and 8q24 genes with Pca in Asian populations. Further work is still required to determine the functional variations and finally clarify the underlying biological mechanisms.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/genética , Receptores Acoplados a Proteínas G/genética , Factores de Transcripción/genética , Adulto , Anciano , Alelos , Cromosomas Humanos Par 8 , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/patología , Factores de Transcripción del Factor Regulador X , Riesgo
8.
PLoS One ; 10(7): e0131763, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26134406

RESUMEN

We have recently reported that ROCK1 deficiency in mouse embryonic fibroblasts (MEF) has superior anti-apoptotic and pro-survival effects than antioxidants against doxorubicin, a chemotherapeutic drug. Although oxidative stress is the most widely accepted mechanism, our studies suggest that ROCK1-dependent actin cytoskeleton remodeling plays a more important role in mediating doxorubicin cytotoxicity on MEFs. To further explore the contributions of ROCK1-dependent actin cytoskeleton remodeling in response to stress, this study investigates the mechanistic differences between the cytotoxic effects of doxorubicin versus hydrogen peroxide (H2O2), with a focus on cytoskeleton alterations, apoptosis and necrosis induction. We found that both types of stress induce caspase activation but with different temporal patterns and magnitudes in MEFs: H2O2 induces the maximal levels (2 to 4-fold) of activation of caspases 3, 8, and 9 within 4 h, while doxorubicin induces much higher maximal levels (15 to 25-fold) of caspases activation at later time points (16-24 h). In addition, necrosis induced by H2O2 reaches maximal levels within 4 h while doxorubicin-induced necrosis largely occurs at 16-24 h secondary to apoptosis. Moreover, both types of stress induce actin cytoskeleton remodeling but with different characteristics: H2O2 induces disruption of stress fibers associated with cytosolic translocation of phosphorylated myosin light chain (p-MLC) from stress fibers, while doxorubicin induces cortical F-actin formation associated with cortical translocation of p-MLC from central stress fibers. Furthermore, N-acetylcysteine (an antioxidant) is a potent suppressor for H2O2-induced cytotoxic effects including caspase activation, necrosis, and cell detachment, but shows a much reduced inhibition on doxorubicin-induced changes. On the other hand, ROCK1 deficiency is a more potent suppressor for the cytotoxic effects induced by doxorubicin than by H2O2. These results support the notion that doxorubicin induces caspase activation, necrosis, and actin cytoskeleton alterations largely through ROCK1-dependent and oxidative stress-independent pathways.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Doxorrubicina/química , Estrés Oxidativo , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos/química , Antioxidantes/metabolismo , Apoptosis , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Supervivencia Celular , Activación Enzimática , Fibroblastos/metabolismo , Peróxido de Hidrógeno/química , Ratones , Microscopía Fluorescente , Necrosis , Fosforilación , Fibras de Estrés/patología
9.
PLoS One ; 9(3): e90758, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24595357

RESUMEN

We have recently reported that the homologous Rho kinases, ROCK1 and ROCK2, play different roles in regulating stress-induced stress fiber disassembly and cell detachment, and the ROCK1 deficiency in mouse embryonic fibroblasts (MEF) has remarkable anti-apoptotic, anti-detachment and pro-survival effects against doxorubicin, a chemotherapeutic drug. This study investigated the roles of ROCK isoforms in doxorubicin-induced reactive oxygen species (ROS) generation which is believed to be the major mechanism underlying its cytotoxicity to normal cells, and especially to cardiomyocytes. Different antioxidants have been shown to provide a protective role reported in numerous experimental studies, but clinical trials of antioxidant therapy showed insufficient benefit against the cardiac side effect. We found that both ROCK1-/- and ROCK2-/- MEFs exhibited reduced ROS production in response to doxorubicin treatment. Interestingly, only ROCK1 deficiency, but not ROCK2 deficiency, significantly enhanced the protective effects of antioxidants against doxorubicin-induced cytotoxicity. First, ROCK1 deficiency and N-acetylcysteine (an anti-oxidant) treatment synergistically reduced ROS levels, caspase activation and cell detachment. In addition, the reduction of ROS generation in ROCK1-/- MEFs in response to doxorubicin treatment was in part through inhibiting NADPH oxidase activity. Furthermore, ROCK1 deficiency enhanced the inhibitory effects of diphenyleneiodonium (an inhibitor of NADPH oxidase) on ROS generation and caspase 3 activation induced by doxorubicin. Finally, ROCK1 deficiency had greater protective effects than antioxidant treatment, especially on reducing actin cytoskeleton remodeling. ROCK1 deficiency not only reduced actomyosin contraction but also preserved central stress fiber stability, whereas antioxidant treatment only reduced actomyosin contraction without preserving central stress fibers. These results reveal a novel strategy to enhance the protective effect of antioxidant therapy by targeting the ROCK1 pathway to stabilize the actin cytoskeleton and boost the inhibitory effects on ROS production, apoptosis and cell detachment.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/efectos adversos , Fibroblastos/efectos de los fármacos , Eliminación de Gen , Quinasas Asociadas a rho/genética , Animales , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
10.
Cell Cycle ; 12(10): 1492-500, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23598717

RESUMEN

The homologous Rho kinases, ROCK1 and ROCK2, are involved in stress fiber assembly and cell adhesion and are assumed to be functionally redundant. Using mouse embryonic fibroblasts (MEFs) derived from ROCK1(-/-) and ROCK2(-/-) mice, we have recently reported that they play different roles in regulating doxorubicin-induced stress fiber disassembly and cell detachment: ROCK1 is involved in destabilizing the actin cytoskeleton and cell detachment, whereas ROCK2 is required for stabilizing the actin cytoskeleton and cell adhesion. Here, we present additional insights into the roles of ROCK1 and ROCK2 in regulating stress-induced impairment of cell-matrix and cell-cell adhesion. In response to doxorubicin, ROCK1(-/-) MEFs showed significant preservation of both focal adhesions and adherens junctions, while ROCK2(-/-) MEFs exhibited impaired focal adhesions but preserved adherens junctions compared with the wild-type MEFs. Additionally, inhibition of focal adhesion or adherens junction formations by chemical inhibitors abolished the anti-detachment effects of ROCK1 deletion. Finally, ROCK1(-/-) MEFs, but not ROCK2(-/-) MEFs, also exhibited preserved central stress fibers and reduced cell detachment in response to serum starvation. These results add new insights into a novel mechanism underlying the anti-detachment effects of ROCK1 deletion mediated by reduced peripheral actomyosin contraction and increased actin stabilization to promote cell-cell and cell-matrix adhesion. Our studies further support the differential roles of ROCK isoforms in regulating stress-induced loss of central stress fibers and focal adhesions as well as cell detachment.


Asunto(s)
Quinasas Asociadas a rho/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Uniones Adherentes/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cofilina 1/metabolismo , Doxorrubicina/farmacología , Ácido Egtácico/farmacología , Adhesiones Focales/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Fibras de Estrés/efectos de los fármacos , Quinasas Asociadas a rho/deficiencia , Quinasas Asociadas a rho/genética
11.
Cell Death Dis ; 4: e483, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23392171

RESUMEN

This study, using mouse embryonic fibroblast (MEF) cells derived from ROCK1(-/-) and ROCK2(-/-) mice, is designed to dissect roles for ROCK1 and ROCK2 in regulating actin cytoskeleton reorganization induced by doxorubicin, a chemotherapeutic drug. ROCK1(-/-) MEFs exhibited improved actin cytoskeleton stability characterized by attenuated periphery actomyosin ring formation and preserved central stress fibers, associated with decreased myosin light chain 2 (MLC2) phosphorylation but preserved cofilin phosphorylation. These effects resulted in a significant reduction in cell shrinkage, detachment, and predetachment apoptosis. In contrast, ROCK2(-/-) MEFs showed increased periphery membrane folding and impaired cell adhesion, associated with reduced phosphorylation of both MLC2 and cofilin. Treatment with inhibitor of myosin (blebbistatin), inhibitor of actin polymerization (cytochalasin D), and ROCK pan-inhibitor (Y27632) confirmed the contributions of actomyosin contraction and stress fiber instability to stress-induced actin cytoskeleton reorganization. These results support a novel concept that ROCK1 is involved in destabilizing actin cytoskeleton through regulating MLC2 phosphorylation and peripheral actomyosin contraction, whereas ROCK2 is required for stabilizing actin cytoskeleton through regulating cofilin phosphorylation. Consequently, ROCK1 and ROCK2 can be functional different in regulating stress-induced stress fiber disassembly and cell detachment.


Asunto(s)
Quinasas Asociadas a rho/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Amidas/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Miosinas Cardíacas/metabolismo , Caspasas/metabolismo , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cofilina 1/metabolismo , Citocalasina D/farmacología , Doxorrubicina/farmacología , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Piridinas/farmacología , Quinasas Asociadas a rho/deficiencia , Quinasas Asociadas a rho/genética
12.
Cell ; 150(3): 575-89, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863010

RESUMEN

The mechanism by which cells decide to skip mitosis to become polyploid is largely undefined. Here we used a high-content image-based screen to identify small-molecule probes that induce polyploidization of megakaryocytic leukemia cells and serve as perturbagens to help understand this process. Our study implicates five networks of kinases that regulate the switch to polyploidy. Moreover, we find that dimethylfasudil (diMF, H-1152P) selectively increased polyploidization, mature cell-surface marker expression, and apoptosis of malignant megakaryocytes. An integrated target identification approach employing proteomic and shRNA screening revealed that a major target of diMF is Aurora kinase A (AURKA). We further find that MLN8237 (Alisertib), a selective inhibitor of AURKA, induced polyploidization and expression of mature megakaryocyte markers in acute megakaryocytic leukemia (AMKL) blasts and displayed potent anti-AMKL activity in vivo. Our findings provide a rationale to support clinical trials of MLN8237 and other inducers of polyploidization and differentiation in AMKL.


Asunto(s)
Azepinas/farmacología , Descubrimiento de Drogas , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Megacariocitos/metabolismo , Poliploidía , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Megacarioblástica Aguda/genética , Megacariocitos/citología , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas Asociadas a rho/metabolismo
13.
Blood ; 120(14): 2868-78, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-22889758

RESUMEN

Erythropoiesis is a dynamic, multistep process whereby hematopoietic stem cells differentiate toward a progressively committed erythroid lineage through intermediate progenitors. Although several downstream signaling molecules have been identified that regulate steady-state erythropoiesis, the major regulators under conditions of stress remain poorly defined. Rho kinases (ROCKs) belong to a family of serine/threonine kinases. Using gene-targeted ROCK1-deficient mice, we show that lack of ROCK1 in phenylhydrazine-induced oxidative stress model results in enhanced recovery from hemolytic anemia as well as enhanced splenic stress erythropoiesis compared with control mice. Deficiency of ROCK1 also results in enhanced survival, whereas wild-type mice die rapidly in response to stress. Enhanced survivability of ROCK1-deficient mice is associated with reduced level of reactive oxygen species. BM transplantation studies revealed that enhanced stress erythropoiesis in ROCK1-deficient mice is stem cell autonomous. We show that ROCK1 binds to p53 and regulates its stability and expression. In the absence of ROCK1, p53 phosphorylation and expression is significantly reduced. Our findings reveal that ROCK1 functions as a physiologic regulator of p53 under conditions of erythroid stress. These findings are expected to offer new perspectives on stress erythropoiesis and may provide a potential therapeutic target in human disease characterized by anemia.


Asunto(s)
Anemia Hemolítica/mortalidad , Anemia Hemolítica/prevención & control , Apoptosis , Eritropoyesis/fisiología , Estrés Oxidativo/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Quinasas Asociadas a rho/fisiología , Anemia Hemolítica/inducido químicamente , Animales , Antimetabolitos Antineoplásicos/toxicidad , Western Blotting , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/patología , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Eritropoyesis/efectos de los fármacos , Eritropoyetina/sangre , Femenino , Citometría de Flujo , Fluorouracilo/toxicidad , Inmunoprecipitación , Masculino , Ratones , Ratones Noqueados , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fenilhidrazinas/toxicidad , Fosforilación , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética
14.
Am J Physiol Heart Circ Physiol ; 302(8): H1603-13, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22328080

RESUMEN

Doxorubicin is a highly effective chemotherapeutic agent used for treating a wide spectrum of tumors, but its usage is limited because of its dose-dependent cardiotoxicity, especially in pediatric patients. Accumulating evidence indicates that caspase-dependent apoptosis contributes to the cardiotoxicity of doxorubicin. However, less attention has been paid to the effects of age on doxorubicin-induced apoptosis signaling in myocardium. This study focused on investigating differential apoptotic sensitivity between neonatal and adult myocardium, in particular, between neonatal and adult cardiomyocytes in vivo. Our results show that caspase-3 activity in normal mouse hearts decreased by ≥ 20-fold within the first 3 wk after birth, associated with a rapid downregulation in the expression of key proapoptotic proteins in intrinsic and extrinsic pathways. This rapid downregulation of caspase-3 activity was confirmed by immunostaining for cleaved caspase-3 and terminal deoxynucleotidyl transferase dUTP-mediated nick-end label staining. Doxorubicin treatment induced a dose-dependent increase in caspase-3 activity and apoptosis in neonatal mouse hearts, and both caspase-8 and caspase-9 activations were involved. Using transgenic mice with a nuclear localized LacZ reporter gene to label cardiomyocytes in vivo, we observed a fourfold higher level of doxorubicin-induced cardiomyocyte apoptosis in 1-wk-old mice compared with that in 3-wk-old mice. This study points to a major difference in apoptotic signaling in doxorubicin cardiotoxicity between neonatal and adult mouse hearts and reveals a critical transition from high to low susceptibility to doxorubicin-induced apoptosis during postnatal heart maturation.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Corazón/crecimiento & desarrollo , Miocardio/citología , Animales , Animales Recién Nacidos/fisiología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Caspasas/fisiología , Regulación hacia Abajo , Activación Enzimática/fisiología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Operón Lac/genética , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/fisiología , Transducción de Señal/fisiología , Fracciones Subcelulares/fisiología
15.
Cancer Cell ; 20(3): 357-69, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21907926

RESUMEN

We show constitutive activation of Rho kinase (ROCK) in cells bearing oncogenic forms of KIT, FLT3, and BCR-ABL, which is dependent on PI3K and Rho GTPase. Genetic or pharmacologic inhibition of ROCK in oncogene-bearing cells impaired their growth as well as the growth of acute myeloid leukemia patient-derived blasts and prolonged the life span of mice bearing myeloproliferative disease. Downstream from ROCK, rapid dephosphorylation or loss of expression of myosin light chain resulted in enhanced apoptosis, reduced growth, and loss of actin polymerization in oncogene-bearing cells leading to significantly prolonged life span of leukemic mice. In summary we describe a pathway involving PI3K/Rho/ROCK/MLC that may contribute to myeloproliferative disease and/or acute myeloid leukemia in humans.


Asunto(s)
Transformación Celular Neoplásica , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia/metabolismo , Trastornos Mieloproliferativos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor de Células Madre/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Animales , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Proteínas de Fusión bcr-abl/biosíntesis , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia/mortalidad , Leucemia/patología , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/mortalidad , Trastornos Mieloproliferativos/patología , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosforilación , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética , Factor de Células Madre/biosíntesis , Factor de Células Madre/genética , Tirosina Quinasa 3 Similar a fms/biosíntesis , Tirosina Quinasa 3 Similar a fms/genética , Proteínas de Unión al GTP rho/biosíntesis , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
16.
J Mol Cell Cardiol ; 49(5): 819-28, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20709073

RESUMEN

Pathological cardiac hypertrophy caused by diverse etiologies eventually leads to cardiac dilation and functional decompensation. We have recently reported that genetic deletion of Rho-associated coiled-coil containing protein kinase 1 (ROCK1) inhibited several pathological events including cardiomyocyte apoptosis in compensated hypertrophic hearts. The present study investigated whether ROCK1 deficiency can prevent the transition from hypertrophy to heart failure. Transgenic mice with cardiac-restricted overexpression of Gαq develop compensated cardiac hypertrophy at young ages, but progress into lethal cardiomyopathy accompanied by increased apoptosis after pregnancy or at old ages. The studies were first carried out using age- and pregnancy-matched wild-type, Gαq, ROCK1(-/-), and Gαq/ROCK1(-/-) mice. The potent beneficial effect of ROCK1 deletion is demonstrated by abolishment of peripartum mortality, and significant attenuation of left ventricular (LV) dilation, wall thinning, and contractile dysfunction in the peripartum Gαq transgenic mice. Increase in cardiomyocyte apoptosis was suppressed by ROCK1 deletion, associated with increased extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) activation and inhibition of mitochondrial translocation of Bax. In addition, ROCK1 deficiency also improved survival, inhibited cardiomyocyte apoptosis, and preserved LV dimension and function in old Gαq mice at 12 months. Furthermore, transgenic overexpression of ROCK1 increased cardiomyocyte apoptosis and accelerated hypertrophic decompensation in Gαq hearts in the absence of pregnancy stress. The present study provides for the first time in vivo evidence for the long-term beneficial effects of ROCK1 deficiency in hypertrophic decompensation and suggests that ROCK1 may be an attractive therapeutic target to limit heart failure progression.


Asunto(s)
Cardiomegalia/enzimología , Progresión de la Enfermedad , Insuficiencia Cardíaca/enzimología , Quinasas Asociadas a rho/metabolismo , Envejecimiento/patología , Animales , Apoptosis , Cardiomegalia/fisiopatología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Eliminación de Gen , Insuficiencia Cardíaca/fisiopatología , Pruebas de Función Cardíaca , Ratones , Miocardio/enzimología , Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Especificidad de Órganos , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo
17.
Blood ; 115(9): 1785-96, 2010 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-20008297

RESUMEN

Rho kinases belong to a family of serine/threonine kinases whose role in recruitment and migration of inflammatory cells is poorly understood. We show that deficiency of ROCK1 results in increased recruitment and migration of macrophages and neutrophils in vitro and in vivo. Enhanced migration resulting from ROCK1 deficiency is observed despite normal expression of ROCK2 and a significant reduction in overall ROCK activity. ROCK1 directly binds PTEN in response to receptor activation and is essential for PTEN phosphorylation and stability. In the absence of ROCK1, PTEN phosphorylation, stability, and its activity are significantly impaired. Consequently, increased activation of downstream targets of PTEN, including PIP3, AKT, GSK-3beta, and cyclin D1, is observed. Our results reveal ROCK1 as a physiologic regulator of PTEN whose function is to repress excessive recruitment of macrophages and neutrophils during acute inflammation.


Asunto(s)
Macrófagos/fisiología , Neutrófilos/fisiología , Fosfohidrolasa PTEN/metabolismo , Quinasas Asociadas a rho/fisiología , Actinas/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Técnicas In Vitro , Inflamación/patología , Inflamación/fisiopatología , Ratones , Ratones Noqueados , Modelos Biológicos , Fosfohidrolasa PTEN/química , Peritonitis/patología , Peritonitis/fisiopatología , Fosforilación , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cicatrización de Heridas/fisiología , Quinasas Asociadas a rho/deficiencia , Quinasas Asociadas a rho/genética
18.
Arch Immunol Ther Exp (Warsz) ; 57(6): 435-45, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19866340

RESUMEN

Doxorubicin (DOX) is one of the most widely used and successful antitumor drugs, but its cumulative and dose-dependent cardiac toxicity has been a major concern of oncologists in cancer therapeutic practice for decades. With the increasing population of cancer survivors, there is a growing need to develop preventive strategies and effective therapies against DOX-induced cardiotoxicity, in particular late-onset cardiomyopathy. Although intensive investigations on DOX-induced cardiotoxicity have continued for decades, the underlying mechanisms responsible for DOX-induced cardiotoxicity have not been completely elucidated. A rapidly expanding body of evidence supports the notion that cardiomyocyte death by apoptosis and necrosis is a primary mechanism of DOX-induced cardiomyopathy and that other types of cell death, such as autophagy and senescence/aging, may participate in this process. This review focuses on the current understanding of the molecular mechanisms underlying DOX-induced cardiomyocyte death, including the major primary mechanism of excess production of reactive oxygen species (ROS) and other recently discovered ROS-independent mechanisms. The different sensitivities to DOX-induced cell death signals between adult and young cardiomyocytes will also be discussed.


Asunto(s)
Cardiomiopatías/inducido químicamente , Cardiotoxinas/efectos adversos , Doxorrubicina/efectos adversos , Neoplasias/tratamiento farmacológico , Animales , Cardiomiopatías/prevención & control , Cardiotoxinas/uso terapéutico , Muerte Celular/efectos de los fármacos , Senescencia Celular , Doxorrubicina/uso terapéutico , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno
19.
J Biol Chem ; 284(18): 11776-80, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19276091

RESUMEN

Insulin signaling is essential for normal glucose homeostasis. Rho-kinase (ROCK) isoforms have been shown to participate in insulin signaling and glucose metabolism in cultured cell lines. To investigate the physiological role of ROCK1 in the regulation of whole body glucose homeostasis and insulin sensitivity in vivo, we studied mice with global disruption of ROCK1. Here we show that, at 16-18 weeks of age, ROCK1-deficient mice exhibited insulin resistance, as revealed by the failure of blood glucose levels to decrease after insulin injection. However, glucose tolerance was normal in the absence of ROCK1. These effects were independent of changes in adiposity. Interestingly, ROCK1 gene ablation caused a significant increase in glucose-induced insulin secretion, leading to hyperinsulinemia. To determine the mechanism(s) by which deletion of ROCK1 causes insulin resistance, we measured the ability of insulin to activate phosphatidylinositol 3-kinase and multiple distal pathways in skeletal muscle. Insulin-stimulated phosphatidylinositol 3-kinase activity associated with IRS-1 or phospho-tyrosine was also reduced approximately 40% without any alteration in tyrosine phosphorylation of insulin receptor in skeletal muscle. Concurrently, serine phosphorylation of IRS-1 at serine 632/635, which is phosphorylated by ROCK in vitro, was also impaired in these mice. Insulin-induced phosphorylation of Akt, AS160, S6K, and S6 was also decreased in skeletal muscle. These data suggest that ROCK1 deficiency causes systemic insulin resistance by impairing insulin signaling in skeletal muscle. Thus, our results identify ROCK1 as a novel regulator of glucose homeostasis and insulin sensitivity in vivo, which could lead to new treatment approaches for obesity and type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Transducción de Señal , Adiposidad/genética , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Glucosa/genética , Insulina/genética , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/terapia , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Quinasas Asociadas a rho
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA