Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Mol Cell Cardiol ; 109: 38-47, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28684310

RESUMEN

AIMS: Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS: We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS: Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Membrana Celular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Ácido Aurintricarboxílico/farmacología , ATPasas Transportadoras de Calcio/genética , Membrana Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
2.
Arterioscler Thromb Vasc Biol ; 34(10): 2310-20, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25147342

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF) has been identified as a crucial regulator of physiological and pathological angiogenesis. Among the intracellular signaling pathways triggered by VEGF, activation of the calcineurin/nuclear factor of activated T cells (NFAT) signaling axis has emerged as a critical mediator of angiogenic processes. We and others previously reported a novel role for the plasma membrane calcium ATPase (PMCA) as an endogenous inhibitor of the calcineurin/NFAT pathway, via interaction with calcineurin, in cardiomyocytes and breast cancer cells. However, the functional significance of the PMCA/calcineurin interaction in endothelial pathophysiology has not been addressed thus far. APPROACH AND RESULTS: Using in vitro and in vivo assays, we here demonstrate that the interaction between PMCA4 and calcineurin in VEGF-stimulated endothelial cells leads to downregulation of the calcineurin/NFAT pathway and to a significant reduction in the subsequent expression of the NFAT-dependent, VEGF-activated, proangiogenic genes RCAN1.4 and Cox-2. PMCA4-dependent inhibition of calcineurin signaling translates into a reduction in endothelial cell motility and blood vessel formation that ultimately impairs in vivo angiogenesis by VEGF. CONCLUSIONS: Given the importance of the calcineurin/NFAT pathway in the regulation of pathological angiogenesis, targeted modulation of PMCA4 functionality might open novel therapeutic avenues to promote or attenuate new vessel formation in diseases that occur with angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Calcineurina/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Proteínas de Unión al Calcio , ATPasas Transportadoras de Calcio/deficiencia , ATPasas Transportadoras de Calcio/genética , Movimiento Celular , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células HEK293 , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isquemia/enzimología , Isquemia/fisiopatología , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
3.
FEBS J ; 286(1): 46-65, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30548183

RESUMEN

Previous studies have demonstrated that activation of calcineurin induces pathological cardiac hypertrophy (CH). In these studies, loss-of-function was mostly achieved by systemic administration of the calcineurin inhibitor cyclosporin A. The lack of conditional knockout models for calcineurin function has impeded progress toward defining the role of this protein during the onset and the development of CH in adults. Here, we exploited a mouse model of CH based on the infusion of a hypertensive dose of angiotensin II (AngII) to model the role of calcineurin in CH in adulthood. AngII-induced CH in adult mice was reduced by treatment with cyclosporin A, without affecting the associated increase in blood pressure, and also by induction of calcineurin deletion in adult mouse cardiomyocytes, indicating that cardiomyocyte calcineurin is required for AngII-induced CH. Surprisingly, cardiac-specific deletion of calcineurin, but not treatment of mice with cyclosporin A, significantly reduced AngII-induced cardiac fibrosis and apoptosis. Analysis of profibrotic genes revealed that AngII-induced expression of Tgfß family members and Lox was not inhibited by cyclosporin A but was markedly reduced by cardiac-specific calcineurin deletion. These results show that AngII induces a direct, calcineurin-dependent prohypertrophic effect in cardiomyocytes, as well as a systemic hypertensive effect that is independent of calcineurin activity.


Asunto(s)
Calcineurina/fisiología , Cardiomegalia/patología , Fibrosis/patología , Miocitos Cardíacos/patología , Angiotensina II/toxicidad , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Progresión de la Enfermedad , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Transducción de Señal , Vasoconstrictores/toxicidad
4.
Oncol Lett ; 16(6): 6950-6959, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30546427

RESUMEN

The present study examined the mRNA expression levels of different isoforms of the plasma membrane calcium ATPase 2 (PMCA2) gene generated by alternative splicing at the first intracellular loop (site A) and C-terminal region (site C) in 85 human breast cancer tumor and 69 adjacent non-tumor tissues. Associations were identified between the expression of PMCA2 splice isoforms and the following clinical variables: Estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status, tumor size, staging and histological classification, and lymph node status. Transcripts including splice site A or splice site C were amplified by reverse transcription-quantitative polymerase chain reaction using PMCA2 isoform-specific primers. Tumor and adjacent tissues were determined to express the different PMCA2 splice isoforms 2w, 2× and 2z (site A), and 2b (site C). The mRNA levels for these variants indicated high biological variability, but increased expression was observed in breast tumor tissues, compared with in adjacent tissues. Significantly increased PMCA2×/b expression levels were detected in breast tumor tissues histologically classified as lobulillar, compared with in ductal-types breast tumor tissues (P<0.028). Furthermore, PMCA2z expression was significantly associated with PR status (P<0.024, compared with in PR-negative tumor tissues), and PMCA2w expression was significantly associated with ER status (P<0.048, increased in ER-positive tumor tissues, compared with ER-negative tumor tissues). Finally, PMCA2b was overexpressed in HER2-positive tumor tissues, compared with in HER2-negative tumor tissues (P<0.014). The data demonstrated the differential mRNA expression of a number of splice site A and C variants of PMCA2 in breast tumor and adjacent tissues, depending on tumor hormone receptor status and histological classification. In agreement with previous data, PMCA2b was overexpressed in HER2-positive tumor tissues, indicating that high mRNA levels of this variant could be a marker of poor prognosis.

5.
Mol Cell Biol ; 35(19): 3409-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26217013

RESUMEN

Emerging evidence indicates that the metalloproteinase Adamts-1 plays a significant role in the pathophysiology of vessel remodeling, but little is known about the signaling pathways that control Adamts-1 expression. We show that vascular endothelial growth factor (VEGF), angiotensin-II, interleukin-1ß, and tumor necrosis factor α, stimuli implicated in pathological vascular remodeling, increase Adamts-1 expression in endothelial and vascular smooth muscle cells. Analysis of the intracellular signaling pathways implicated in this process revealed that VEGF and angiotensin-II upregulate Adamts-1 expression via activation of differential signaling pathways that ultimately promote functional binding of the NFAT or C/EBPß transcription factors, respectively, to the Adamts-1 promoter. Infusion of mice with angiotensin-II triggered phosphorylation and nuclear translocation of C/EBPß proteins in aortic cells concomitantly with an increase in the expression of Adamts-1, further underscoring the importance of C/EBPß signaling in angiotensin-II-induced upregulation of Adamts-1. Similarly, VEGF promoted NFAT activation and subsequent Adamts-1 induction in aortic wall in a calcineurin-dependent manner. Our results demonstrate that Adamts-1 upregulation by inducers of pathological vascular remodeling is mediated by specific signal transduction pathways involving NFAT or C/EBPß transcription factors. Targeting of these pathways may prove useful in the treatment of vascular disease.


Asunto(s)
Proteínas ADAM/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Factores de Transcripción NFATC/metabolismo , Remodelación Vascular , Proteínas ADAM/genética , Proteína ADAMTS1 , Animales , Aorta/enzimología , Secuencia de Bases , Calcineurina/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones Noqueados , Datos de Secuencia Molecular , Transducción de Señal , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA