Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Cell Mol Med ; 25(11): 5316-5325, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33942489

RESUMEN

Type 2 diabetes mellitus (T2DM) leads to monocyte dysfunction associated with atherogenesis and defective arteriogenesis. Transforming growth factor (TGF)-ß1, placenta growth factor (PlGF)-1 and vascular endothelial growth factor (VEGF)A play important roles in atherogenesis and arteriogenesis. VEGF-receptor (VEGFR)-mediated monocyte migration is inhibited in T2DM (VEGFA resistance), while TGF-ß1-induced monocyte migration is fully functional. Therefore, we hypothesize that TGF-ß antagonises the VEGFA responses in human monocytes. We demonstrate that monocytes from T2DM patients have an increased migratory response towards low concentrations of TGF-ß1, while PlGF-1/VEGFA responses are mitigated. Mechanistically, this is due to increased expression of type II TGF-ß receptor in monocytes under high-glucose conditions and increased expression of soluble (s)VEGFR1, which is known to interfere with VEGFA signalling. VEGFA resistance in monocytes from T2DM patients can be rescued by either experimental down-regulation of TGF-ß receptor expression in vitro or by functional blocking of TGF-ß signalling using either a TGF-ß receptor kinase inhibitor or a TGF-ß neutralizing antibody. Our data demonstrate that both T2DM and high-glucose potentiate the TGF-ß pathway. TGF-ß signalling impairs VEGFR-mediated responses in T2DM monocytes and in this way contributes to mononuclear cell dysfunction, provide novel insights into T2DM vascular dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Glucosa/efectos adversos , Monocitos/patología , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Transducción de Señal , Edulcorantes/efectos adversos , Factor de Crecimiento Transformador beta1/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
J Cell Mol Med ; 22(11): 5429-5438, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30102472

RESUMEN

Type 2 diabetes mellitus (T2DM) is a cardiovascular risk factor which leads to atherosclerosis, an inflammatory disease characterized by the infiltration of mononuclear cells in the vessel. Bone morphogenetic protein (BMP)-2 is a cytokine which has been recently shown to be elevated in atherosclerosis and T2DM and to contribute to vascular inflammation. However, the role of BMP-2 in the regulation of mononuclear cell function remains to be established. Herein, we demonstrate that BMP-2 induced human monocyte chemotaxis via phosphoinositide 3 kinase and mitogen-activated protein kinases. Inhibition of endogenous BMP-2 signalling, by Noggin or a BMP receptor inhibitor, interfered with monocyte migration. Although BMP-2 expression was increased in monocytes from T2DM patients, it could still stimulate their migration. Furthermore, BMP-2 interfered with their differentiation into M2 macrophages. Finally, BMP-2 both induced the adhesion of monocytes to fibronectin and endothelial cells (ECs), and promoted the adhesive properties of ECs, by increasing expression of adhesion and pro-inflammatory molecules. Our data demonstrate that BMP-2 could exert its pro-inflammatory effects by inducing monocyte migration and adhesiveness to ECs and by interfering with the monocyte differentiation into M2 macrophages. Our findings provide novel insights into the mechanisms by which BMP-2 may contribute to the development of atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Proteína Morfogenética Ósea 2/genética , Diabetes Mellitus Tipo 2/genética , Macrófagos/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas Portadoras/genética , Adhesión Celular/genética , Diferenciación Celular/genética , Quimiotaxis/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Fibronectinas/genética , Regulación de la Expresión Génica/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/patología , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Monocitos/patología , Fosfatidilinositol 3-Quinasa/genética , Transducción de Señal
3.
Int J Mol Sci ; 18(10)2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29039786

RESUMEN

Fibrotic diseases are characterized by net accumulation of extracellular matrix proteins in affected organs leading to their dysfunction and ultimate failure. Myofibroblasts have been identified as the cells responsible for the progression of the fibrotic process, and they originate from several sources, including quiescent tissue fibroblasts, circulating CD34⁺ fibrocytes and the phenotypic conversion of various cell types into activated myofibroblasts. Several studies have demonstrated that endothelial cells can transdifferentiate into mesenchymal cells through a process termed endothelial- mesenchymal transition (EndMT) and that this can give rise to activated myofibroblasts involved in the development of fibrotic diseases. Transforming growth factor ß (TGF-ß) has a central role in fibrogenesis by modulating the fibroblast phenotype and function, inducing myofibroblast transdifferentiation and promoting matrix accumulation. In addition, TGF-ß by inducing EndMT may further contribute to the development of fibrosis. Despite extensive investigation of the pathogenesis of fibrotic diseases, no effective treatment strategies are available. Delineation of the mechanisms responsible for initiation and progression of fibrotic diseases is crucial for the development of therapeutic strategies for the treatment of the disease. In this review, we summarize the role of the TGF-ß signaling pathway and EndMT in the development of fibrotic diseases and discuss their therapeutic potential.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibrosis/etiología , Fibrosis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biomarcadores , Transdiferenciación Celular , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis/diagnóstico , Humanos , Miofibroblastos/metabolismo , Transducción de Señal
4.
J Biol Chem ; 287(22): 18551-61, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22493445

RESUMEN

Genetic and molecular studies suggest that activin receptor-like kinase 1 (ALK1), a transforming growth factor ß (TGF-ß) type I receptor, and endoglin, a TGF-ß co-receptor, play an essential role in vascular development and pathological angiogenesis. Several agents that interfere with ALK1 and endoglin function are currently in clinical trials for antiangiogenic activity in cancer therapy. One of these agents, PF-03446962 (anti-hALK1 antibody), shows promising results in the clinic. However, its effects on endothelial cell function and mechanism of action are unclear. Here we demonstrate that anti-hALK1 antibody selectively recognizes human ALK1. The anti-hALK1 antibody interfered with bone morphogenetic protein 9 (BMP9)-induced signaling in endothelial cells. Consistent with this notion, anti-hALK1 antibody was found to compete highly efficiently with the binding of the ALK1 ligand BMP9 and TGF-ß to ALK1. Moreover, it prevented BMP9-dependent recruitment of co-receptor endoglin into this angiogenesis-mediating signaling complex. In addition, we demonstrated that anti-hALK1 antibody inhibited endothelial cell sprouting but did not directly interfere with vascular endothelial growth factor (VEGF) signaling, VEGF-induced proliferation, and migration of endothelial cells. Finally, we demonstrated that BMP9 in serum is essential for endothelial sprouting and that anti-hALK1 antibody inhibits this potently. Our data suggest that both the VEGF/VEGF receptor and the BMP9/ALK1 pathways are essential for stimulating angiogenesis, and targeting both pathways simultaneously may be an attractive strategy to overcome resistance to antiangiogenesis therapy.


Asunto(s)
Receptores de Activinas Tipo II/inmunología , Endotelio Vascular/metabolismo , Factor 2 de Diferenciación de Crecimiento/fisiología , Transducción de Señal/fisiología , Receptores de Activinas Tipo II/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Citometría de Flujo , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , Unión Proteica
5.
Proc Natl Acad Sci U S A ; 107(14): 6340-5, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308542

RESUMEN

Annexin A1 (AnxA1) is a candidate regulator of the epithelial- to mesenchymal (EMT)-like phenotypic switch, a pivotal event in breast cancer progression. We show here that AnxA1 expression is associated with a highly invasive basal-like breast cancer subtype both in a panel of human breast cancer cell lines as in breast cancer patients and that AnxA1 is functionally related to breast cancer progression. AnxA1 knockdown in invasive basal-like breast cancer cells reduced the number of spontaneous lung metastasis, whereas additional expression of AnxA1 enhanced metastatic spread. AnxA1 promotes metastasis formation by enhancing TGFbeta/Smad signaling and actin reorganization, which facilitates an EMT-like switch, thereby allowing efficient cell migration and invasion of metastatic breast cancer cells.


Asunto(s)
Anexina A1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Anexina A1/genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Trasplante de Neoplasias
6.
J Cell Mol Med ; 16(10): 2440-50, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22436015

RESUMEN

Transforming growth factor-beta (TGF-ß) stimulates both ischaemia induced angiogenesis and shear stress induced arteriogenesis by signalling through different receptors. How these receptors are involved in both these processes of blood flow recovery is not entirely clear. In this study the role of TGF-ß receptors 1 and endoglin is assessed in neovascularization in mice. Unilateral femoral artery ligation was performed in mice heterozygous for either endoglin or ALK1 and in littermate controls. Compared with littermate controls, blood flow recovery, monitored by laser Doppler perfusion imaging, was significantly hampered by maximal 40% in endoglin heterozygous mice and by maximal 49% in ALK1 heterozygous mice. Collateral artery size was significantly reduced in endoglin heterozygous mice compared with controls but not in ALK1 heterozygous mice. Capillary density in ischaemic calf muscles was unaffected, but capillaries from endoglin and ALK1 heterozygous mice were significantly larger when compared with controls. To provide mechanistic evidence for the differential role of endoglin and ALK1 in shear induced or ischaemia induced neovascularization, murine endothelial cells were exposed to shear stress in vitro. This induced increased levels of endoglin mRNA but not ALK1. In this study it is demonstrated that both endoglin and ALK1 facilitate blood flow recovery. Importantly, endoglin contributes to both shear induced collateral artery growth and to ischaemia induced angiogenesis, whereas ALK1 is only involved in ischaemia induced angiogenesis.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Arterias/crecimiento & desarrollo , Circulación Colateral , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Estrés Fisiológico , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II , Animales , Capilares/metabolismo , Bovinos , Células Cultivadas , Endoglina , Células Endoteliales/metabolismo , Heterocigoto , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Isquemia/genética , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Neovascularización Patológica/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Estrés Mecánico , Regulación hacia Arriba
7.
Mol Cell Proteomics ; 9(9): 1982-90, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20511396

RESUMEN

Genetic and biochemical studies have shown that selective interactions between the Jun, Fos, and activating transcription factor (ATF) components of transcription factor activating protein 1 (AP-1) exhibit specific and critical functions in the regulation of cell proliferation, differentiation, and survival. For instance, the ratio between c-Jun/c-Fos and c-Jun/ATF2 dimers in the cell can be a determining factor in the cellular response to oncogenic or apoptotic stimuli. Until recently, no methods were available to detect endogenous AP-1 complexes in cells and tissues in situ. Here, we validated the proximity ligation assay (PLA) for its ability to specifically visualize and quantify changes in endogenous c-Jun/c-Fos, c-Jun/ATF2, and c-Jun/Fra1 complexes by using, among others, partner-selective c-Jun mutants. Furthermore, we examined the levels of c-Jun/AP-1 dimers in cell lines representing different types of human breast cancer and found that aggressive basal-like breast cancer cells can be discriminated from much less invasive luminal-like cells by PLA detection of c-Jun/Fra1 rather than of c-Jun/ATF2 and c-Jun/c-Fos. Also in tumor tissue derived from highly metastatic basal-like MDA-MB231 cells, high levels of c-Jun/Fra1 complexes were detected. Together, these results demonstrate that in situ PLA is a powerful diagnostic tool to analyze and quantify the amounts of biologically critical AP-1 dimers in fixed cells and tissue material.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/metabolismo , Western Blotting , Neoplasias de la Mama/patología , Línea Celular Tumoral , Dimerización , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Mutación , Proteínas Proto-Oncogénicas c-jun/genética
8.
Int Immunopharmacol ; 105: 108523, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35086057

RESUMEN

PHA-022121 is a novel small molecule bradykinin B2 receptor antagonist, in clinical development for the treatment and prevention of hereditary angioedema attacks. The present study describes the in vitro pharmacological characteristics of PHA-022121 and its active metabolite, PHA-022484 (M2-D). In mammalian cell lines, PHA-022121 and PHA-022484 show high affinity for the recombinant human bradykinin B2 receptor with Ki values of 0.47 and 0.70 nM, respectively, and potent antagonism of the human bradykinin B2 receptor with Kb values of 0.15 and 0.26 nM, respectively (calcium mobilization assay). Antagonist potency at the recombinant cynomolgus monkey bradykinin B2 receptor is similarly high (Kb values of 1.42 and 1.12 nM for PHA-022121 and PHA-022484, respectively), however, potency at rat, mouse, rabbit and dog bradykinin B2 receptors is at least 100-fold lower than the potency at the human receptor for both compounds. In the human umbilical vein contractility assay, both PHA-022121 and PHA-022484 show a potent, surmountable and reversible B2 antagonist activity with pA2 values of 0.35 and 0.47 nM, respectively. The in vitro off-target profile of PHA-022121 and PHA-022484 demonstrates a high degree of selectivity over a wide range of molecular targets, including the bradykinin B1 receptor. It is concluded that PHA-022121 is a novel, low-molecular weight, competitive antagonist of the human bradykinin B2 receptor with high affinity, high antagonist potency, and high selectivity. It is about 20-fold more potent than icatibant at the human bradykinin B2 receptor as assessed using recombinant or endogenously expressed receptors.


Asunto(s)
Antagonistas de los Receptores de Bradiquinina , Bradiquinina , Animales , Unión Competitiva , Bradiquinina/metabolismo , Antagonistas de los Receptores de Bradiquinina/metabolismo , Antagonistas de los Receptores de Bradiquinina/farmacología , Perros , Macaca fascicularis/metabolismo , Mamíferos , Ratones , Conejos , Ratas , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo , Venas Umbilicales/metabolismo
9.
J Cell Mol Med ; 15(12): 2723-34, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21306557

RESUMEN

Previously we observed that cardiomyocyte progenitor cells (hCMPCs) isolated from the human heart differentiate spontaneously into cardiomyocytes and vascular cells when transplanted after myocardial infarction (MI) in the ischemic heart. After MI, deprivation of oxygen is the first major change in the cardiac environment. How cells handle hypoxia is highly cell type dependent. The effect of hypoxia on cardiac stem or progenitor cells remains to be elucidated. Here, we show for the first time that short- and long-term hypoxia have different effects on hCMPCs. Short-term hypoxia increased the migratory and invasive capacities of hCMPCs likely via mesenchymal transformation. Although long-term exposure to low oxygen levels did not induce differentiation of hCMPCs into mature cardiomyocytes or endothelial cells, it did increase their proliferation, stimulated the secretome of the cells which was shifted to a more anti-inflammatory profile and dampened the migration by altering matrix metalloproteinase (MMP) modulators. Interestingly, hypoxia greatly induced the expression of the extracellular matrix modulator thrombospondin-2 (TSP-2). Knockdown of TSP-2 resulted in increased proliferation, migration and MMP activity. In conclusion, short exposure to hypoxia increases migratory and invasive capacities of hCMPCs and prolonged exposure induces proliferation, an angiogenic secretion profile and dampens migration, likely controlled by TSP-2.


Asunto(s)
Hipoxia de la Célula , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Células Madre/metabolismo , Western Blotting , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Feto/citología , Feto/metabolismo , Regulación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Metaloproteinasas de la Matriz/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Trombospondina 1/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/metabolismo
10.
J Cell Sci ; 122(Pt 18): 3294-302, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706683

RESUMEN

Vascular endothelial growth factor (VEGF) and transforming growth factor-beta (TGFbeta) are potent regulators of angiogenesis. How VEGF and TGFbeta signaling pathways crosstalk is not well understood. Therefore, we analyzed the effects of the TGFbeta type-I-receptor inhibitors (SB-431542 and LY-2157299) and VEGF on endothelial cell (EC) function and angiogenesis. We show that SB-431542 dramatically enhances VEGF-induced formation of EC sheets from fetal mouse metatarsals. Sub-optimal doses of VEGF and SB-431542 synergistically induced EC migration and sprouting of EC spheroids, whereas overexpression of a constitutively active form of TGFbeta type-I receptor had opposite effects. Using quantitative PCR, we demonstrated that VEGF and SB-431542 synergistically upregulated the mRNA expression of genes involved in angiogenesis, including the integrins alpha5 and beta3. Specific downregulation of alpha5-integrin expression or functional blocking of alpha5 integrin with a specific neutralizing antibody inhibited the cooperative effect of VEGF and SB-431542 on EC sprouting. In vivo, LY-2157299 induced angiogenesis and enhanced VEGF- and basic-fibroblast-growth-factor-induced angiogenesis in a Matrigel-plug assay, whereas adding an alpha5-integrin-neutralizing antibody to the Matrigel selectively inhibited this enhanced response. Thus, induction of alpha5-integrin expression is a key determinant by which inhibitors of TGFbeta type-I receptor kinase and VEGF synergistically promote angiogenesis.


Asunto(s)
Integrina alfa5/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Benzamidas , Bioensayo , Movimiento Celular/efectos de los fármacos , Colágeno/metabolismo , Dioxoles , Combinación de Medicamentos , Sinergismo Farmacológico , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Feto/irrigación sanguínea , Feto/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Integrina beta3/metabolismo , Laminina/metabolismo , Ratones , Pruebas de Neutralización , Proteoglicanos/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
11.
Breast Cancer Res Treat ; 128(3): 657-66, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20821046

RESUMEN

Transforming growth factor-ß (TGF-ß) has opposing roles in breast cancer progression by acting as a tumor suppressor in the initial phase, but stimulating invasion and metastasis at later stages. In contrast to the mechanisms by which TGF-ß induces growth arrest, the pathways that mediate tumor invasion are not well understood. Here, we describe a TGF-ß-dependent invasion assay system consisting of spheroids of MCF10A1 normal breast epithelial cells (M1) and RAS-transformed (pre-)malignant derivatives (M2 and M4) embedded in collagen gels. Both basal and TGF-ß-induced invasion of these cell lines was found to correlate with their tumorigenic potential; M4 showing the most aggressive behavior and M1 showing the least. Basal invasion was strongly inhibited by the TGF-ß receptor kinase inhibitor SB-431542, indicating the involvement of autocrine TGF-ß or TGF-ß-like activity. TGF-ß-induced invasion in premalignant M2 and highly malignant M4 cells was also inhibited upon specific knockdown of Smad3 or Smad4. Interestingly, both a broad spectrum matrix metalloproteinase (MMP) inhibitor and a selective MMP2 and MMP9 inhibitor mitigated TGF-ß-induced invasion of M4 cells, while leaving basal invasion intact. In line with this, TGF-ß was found to strongly induce MMP2 and MMP9 expression in a Smad3- and Smad4-dependent manner. This collagen-embedded spheroid system therefore offers a valuable screening model for TGF-ß/Smad- and MMP2- and MMP9-dependent breast cancer invasion.


Asunto(s)
Neoplasias de la Mama , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Invasividad Neoplásica/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Benzamidas/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Dioxoles/farmacología , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica/prevención & control , Proteínas Smad/genética , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Células Tumorales Cultivadas , Regulación hacia Arriba/genética
12.
ESC Heart Fail ; 8(3): 1873-1884, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33779075

RESUMEN

AIMS: Somatic mutations in haematopoietic stem cells can lead to the clonal expansion of mutated blood cells, known as clonal haematopoiesis (CH). Mutations in the most prevalent driver genes DNMT3A and TET2 with a variant allele frequency (VAF) ≥ 2% have been associated with atherosclerosis and chronic heart failure of ischemic origin (CHF). However, the effects of mutations in other driver genes for CH with low VAF (<2%) on CHF are still unknown. METHODS AND RESULTS: Therefore, we analysed mononuclear bone marrow and blood cells from 399 CHF patients by deep error-corrected targeted sequencing of 56 genes and associated mutations with the long-term mortality in these patients (3.95 years median follow-up). We detected 1113 mutations with a VAF ≥ 0.5% in 347 of 399 patients, and only 13% had no detectable CH. Despite a high prevalence of mutations in the most frequently mutated genes DNMT3A (165 patients) and TET2 (107 patients), mutations in CBL, CEBPA, EZH2, GNB1, PHF6, SMC1A, and SRSF2 were associated with increased death compared with the average death rate of all patients. To avoid confounding effects, we excluded patients with DNMT3A-related, TET2-related, and other clonal haematopoiesis of indeterminate potential (CHIP)-related mutations with a VAF ≥ 2% for further analyses. Kaplan-Meier survival analyses revealed a significantly higher mortality in patients with mutations in either of the seven genes (53 patients), combined as the CH-risk gene set for CHF. Baseline patient characteristics showed no significant differences in any parameter including patient age, confounding diseases, severity of CHF, or blood cell parameters except for a reduced number of platelets in patients with mutations in the risk gene set in comparison with patients without. However, carrying a mutation in any of the risk genes remained significant after multivariate cox regression analysis (hazard ratio, 3.1; 95% confidence interval, 1.8-5.4; P < 0.001), whereas platelet numbers did not. CONCLUSIONS: Somatic mutations with low VAF in a distinct set of genes, namely, in CBL, CEBPA, EZH2, GNB1, PHF6, SMC1A, and SRSF2, are significantly associated with mortality in CHF, independently of the most prevalent CHIP-mutations in DNMT3A and TET2. Mutations in these genes are prevalent in young CHF patients and comprise an independent risk factor for the outcome of CHF, potentially providing a novel tool for risk assessment in CHF.


Asunto(s)
Hematopoyesis Clonal , Insuficiencia Cardíaca , Proteínas de Unión al ADN/genética , Insuficiencia Cardíaca/genética , Humanos , Mutación , Proteínas Proto-Oncogénicas/genética
13.
Exp Hematol ; 83: 95-104, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31891750

RESUMEN

Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. Many studies have provided evidence that both genetic and environmental factors induce atherosclerosis, leading thus to cardiovascular complications. Atherosclerosis is an inflammatory disease, and aging is strongly associated with the development of atherosclerosis. Recent experimental evidence suggests that clonal hematopoiesis (CH) is an emerging cardiovascular risk factor that contributes to the development of atherosclerosis and cardiac dysfunction and exacerbates cardiovascular diseases. CH is caused by somatic mutations in recurrent genes in hematopoietic stem cells, leading to the clonal expansion of mutated blood cell clones. Many of the mutated genes are known in the context of myeloid neoplasms. However, only some individuals carrying CH mutations develop hematologic abnormalities. CH is clearly age dependent and is not rare: at least 10%-20% of people >70 years old carry CH. The newly discovered association between myeloid leukemia-driver mutations and the progression of CVDs has raised medical interest. In this review, we summarize the current view on the contribution of CH in different cardiovascular diseases, CVD risk assessment, patient stratification, and the development of novel therapeutic strategies.


Asunto(s)
Envejecimiento , Aterosclerosis , Evolución Clonal/genética , Insuficiencia Cardíaca , Hematopoyesis/genética , Mutación , Accidente Cerebrovascular , Factores de Edad , Anciano , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Factores de Riesgo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
14.
Arterioscler Thromb Vasc Biol ; 27(3): 532-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17194892

RESUMEN

OBJECTIVE: The flow-responsive Kruppel-like factor 2 (KLF2) is crucial for maintaining endothelial cell quiescence. Here, we describe its detailed effects on transforming growth factor-beta (TGF-beta) signaling, which normally has proatherogenic effects on endothelium. METHODS AND RESULTS: In-depth analysis of genome-wide expression data shows that prolonged lentiviral-mediated overexpression of KLF2 in human umbilical vein endothelial cells (HUVECs) diminishes the expression of a large panel of established TGF-beta-inducible genes. Both baseline and TGF-beta-induced expression levels of plasminogen activator inhibitor 1 (PAI-1) and thrombospondin-1 are greatly diminished by KLF2. Using a combination of ectopic expression, small interfering RNA-mediated knockdown, and promoter activity assays, we show that KLF2 partly inhibits the phosphorylation and subsequent nuclear accumulation of Smad2, thereby suppressing the TGF-beta-induced Smad4-mediated transcriptional activity. This is achieved through TGF-beta-independent induction of inhibitory Smad7. Additionally, a full inhibition of TGF-beta signaling is functionally achieved through a simultaneous suppression of activator protein 1 (AP-1), which is an essential cofactor for TGF-beta-dependent transcription of many genes. CONCLUSIONS: The concerted mechanism by which KLF2 inhibits TGF-beta signaling through induction of inhibitory Smad7 and attenuation of AP-1 activity provides a novel mechanism by which KLF2 contributes to sustaining a quiescent, atheroprotective status of vascular endothelium.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Transducción de Señal/fisiología , Proteína smad7/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Western Blotting , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/farmacología , Fosforilación , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteína smad7/genética , Factor de Transcripción AP-1/genética , Venas Umbilicales/citología
15.
Int J Cardiol ; 255: 160-165, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29425557

RESUMEN

BACKGROUND: Hypercholesterolemia (HC) is an important cardiovascular risk factor characterized by elevated low density lipoprotein-cholesterol (LDL-C) plasma levels. HC negatively affects monocyte function by reducing their chemotactic response towards different growth factors. We aimed to elucidate the molecular mechanisms by which LDL induces monocyte dysfunction. METHODS AND RESULTS: Human monocytes exposed to either native (nLDL) or oxidized LDL (oxLDL) in vitro showed reduced chemotactic responses towards vascular endothelial growth factor A (VEGFA) and monocyte chemotactic protein-1 (MCP-1), but displayed enhanced random migration (chemokinesis). Mechanistically, the exposure to LDL resulted in the activation of p38 mitogen-activated protein kinase (MAPK) and modulated MCP-1 and VEGFA-induced signaling in human monocytes. Furthermore, the aberrant p38 activation induced by oxLDL is due to the functional impairment of Dual Specificity Phosphatase-1 (DUSP-1). In the absence of LDL, the pharmacological inhibition of DUSP-1 alone was sufficient to recapitulate the accelerated chemokinetic and blunted chemotactic phenotype of monocytes. Finally, p38 MAPK inhibition in monocytes isolated from hyperlipidemic mice prevented the aberrant chemokinetic phenotype. CONCLUSIONS: Our data demonstrate that LDL induces monocyte chemokinesis of human monocytes by inducing mononuclear cell activation through the aberrant modulation of DUSP-1-p38/MAPK signaling axis. Moreover, our findings suggest that MCP-1/VEGFA-induced chemotaxis is reduced by LDL secondary to the impairment of ligand-induced signaling. These findings provide novel insight into hypercholesterolemia-associated vascular dysfunction and its potential involvement in the pathogenesis of atherosclerosis.


Asunto(s)
Quimiocinas , Quimiotaxis/efectos de los fármacos , Líquido Intracelular/efectos de los fármacos , Lipoproteínas LDL/toxicidad , Monocitos/efectos de los fármacos , Animales , Células Cultivadas , Quimiocinas/metabolismo , Quimiotaxis/fisiología , Humanos , Líquido Intracelular/metabolismo , Ratones , Ratones Noqueados , Monocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
EJNMMI Res ; 6(1): 77, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27778311

RESUMEN

BACKGROUND: Circulating white blood cells crucially contribute to maintenance and repair of solid organs. Therefore, certain cell populations such as monocytes are attractive targets for use in molecular imaging and cell imaging, e.g. after labelling with radionuclides, as well as for cell therapies. However, the preparation of monocytes may require freezing and thawing to preserve cells for timely and standardised applications. Additional modifications of these cells such as radioisotope labelling are necessary prior to their application in vivo. We therefore tested the hypothesis whether cryopreservation of freshly isolated circulating human monocytes affects their functional phenotype or their suitability for radionuclide labelling. RESULTS: CD14+CD16- monocytes were isolated from human peripheral blood. They were either directly used for cellular assays and labelling or frozen down using cryoprotectants. In the latter case, cells were thawed prior to further use and analysed for survival, chemotactic responses to various growth factors and adhesion on endothelial cells. In addition, both fresh and cryopreserved monocytes were labelled with radiotracers followed by assessment of survival and chemotactic responses. In all functional assays performed, cryopreserved monocytes did not significantly differ from freshly isolated monocytes with regard to their functionality. Cryopreservation did not affect cell survival. There was no effect on the chemotactic response of monocytes towards different growth factors. Likewise, adhesion properties remained unchanged following cryopreservation. Moreover, the labelling efficiency was similar for freshly isolated and cryopreserved monocytes. Labelling did not negatively affect monocyte survival and function. CONCLUSIONS: Our data indicate that cryopreservation of freshly isolated human primary monocytes is feasible and does not negatively affect their functionality when used for labelling and functional assessment.

17.
Clin Cancer Res ; 22(1): 96-106, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26373572

RESUMEN

PURPOSE: Antiangiogenic therapy, mostly targeting VEGF, has been applied in cancer patients for the last decade. However, resistance to anti-VEGF therapy and/or no significant benefit as monotherapeutic agent is often observed. Therefore, new antiangiogenic strategies are needed. In the current study, we investigated the therapeutic effect of interfering with the bone morphogenetic protein (BMP)9/activin receptor-like kinase (ALK)1 signaling pathway by using an ALK1-Fc ligand trap. EXPERIMENTAL DESIGN: We analyzed the potential antiangiogenic and antitumor effects of ALK1-Fc protein as monotherapy and in combination with chemotherapy in vivo in mouse models of melanoma, head and neck cancer, and invasive lobular breast carcinomas. ALK1-Fc sequesters BMP9 and 10 and prevents binding of these ligands to endothelial ALK1, which regulates angiogenesis. RESULTS: Treatment of mice with ALK1-Fc strongly decreased the tumors' microvascular density in the three different mouse cancer models. However, this effect was not accompanied by a reduction in tumor volume. An immunohistochemical analysis of the tumor samples revealed that ALK1-Fc treatment increased the pericyte coverage of the remaining tumor vessels and decreased the hypoxia within the tumor. Next, we observed that combining ALK1-Fc with cisplatin inhibited tumor growth in the breast and head and neck cancer models more efficiently than chemotherapy alone. CONCLUSIONS: The addition of ALK1-Fc to the cisplatin treatment was able to enhance the cytotoxic effect of the chemotherapy. Our results provide strong rationale to explore combined targeting of ALK1 with chemotherapy in a clinical setting, especially in the ongoing phase II clinical trials with ALK1-Fc.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Ratones , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neovascularización Patológica/genética , Proteínas Recombinantes de Fusión/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral
18.
PLoS One ; 9(1): e86273, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489709

RESUMEN

ENDOGLIN (ENG) is a co-receptor for transforming growth factor-ß (TGF-ß) family members that is highly expressed in endothelial cells and has a critical function in the development of the vascular system. Mutations in Eng are associated with the vascular disease known as hereditary hemorrhagic telangiectasia type l. Using mouse embryonic stem cells we observed that angiogenic factors, including vascular endothelial growth factor (VEGF), induce vasculogenesis in embryoid bodies even when Eng deficient cells or cells depleted of Eng using shRNA are used. However, ENG is required for the stem cell-derived endothelial cells to organize effectively into tubular structures. Consistent with this finding, fetal metatarsals isolated from E17.5 Eng heterozygous mouse embryos showed reduced VEGF-induced vascular network formation. Moreover, shRNA-mediated depletion and pharmacological inhibition of ENG in human umbilical vein cells mitigated VEGF-induced angiogenesis. In summary, we demonstrate that ENG is required for efficient VEGF-induced angiogenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Endoglina , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Neovascularización Fisiológica/genética
19.
J Exp Med ; 210(3): 563-79, 2013 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-23401487

RESUMEN

Therapy-induced resistance remains a significant hurdle to achieve long-lasting responses and cures in cancer patients. We investigated the long-term consequences of genetically impaired angiogenesis by engineering multiple tumor models deprived of endoglin, a co-receptor for TGF-ß in endothelial cells actively engaged in angiogenesis. Tumors from endoglin-deficient mice adapted to the weakened angiogenic response, and refractoriness to diminished endoglin signaling was accompanied by increased metastatic capability. Mechanistic studies in multiple mouse models of cancer revealed that deficiency for endoglin resulted in a tumor vasculature that displayed hallmarks of endothelial-to-mesenchymal transition, a process of previously unknown significance in cancer biology, but shown by us to be associated with a reduced capacity of the vasculature to avert tumor cell intra- and extravasation. Nevertheless, tumors deprived of endoglin exhibited a delayed onset of resistance to anti-VEGF (vascular endothelial growth factor) agents, illustrating the therapeutic utility of combinatorial targeting of multiple angiogenic pathways for the treatment of cancer.


Asunto(s)
Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Tumores Neuroendocrinos/irrigación sanguínea , Neoplasias Pancreáticas/irrigación sanguínea , Animales , Células Cultivadas , Endoglina , Transición Epitelial-Mesenquimal , Femenino , Proteínas Activadoras de GTPasa/fisiología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Neoplasias Hepáticas Experimentales/secundario , Ratones , Neovascularización Fisiológica , Neoplasias Pancreáticas/patología , Proteína 1 Relacionada con Twist/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
20.
Int J Biol Sci ; 8(2): 195-213, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253564

RESUMEN

Transforming growth factor ß (TGFß) family members are involved in a wide range of diverse functions and play key roles in embryogenesis, development and tissue homeostasis. Perturbation of TGFß signaling may lead to vascular and other diseases. In vitro studies have provided evidence that TGFß family members have a wide range of diverse effects on vascular cells, which are highly dependent on cellular context. Consistent with these observations genetic studies in mice and humans showed that TGFß family members have ambiguous effects on the function of the cardiovascular system. In this review we discuss the recent advances on TGFß signaling in (cardio)vascular diseases, and describe the value of TGFß signaling as both a disease marker and therapeutic target for (cardio)vascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Calcinosis/genética , Calcinosis/metabolismo , Humanos , Ligandos , Ratones , Modelos Biológicos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda