Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carmelo Bernabeu is active.

Publication


Featured researches published by Carmelo Bernabeu.


Clinical Science | 2011

TGF-β/TGF-β receptor system and its role in physiological and pathological conditions.

Juan F. Santibanez; Miguel Quintanilla; Carmelo Bernabeu

The TGF-β (transforming growth factor-β) system signals via protein kinase receptors and Smad mediators to regulate a plethora of biological processes, including morphogenesis, embryonic development, adult stem cell differentiation, immune regulation, wound healing and inflammation. In addition, alterations of specific components of the TGF-β signalling pathway may contribute to a broad range of pathologies such as cancer, cardiovascular pathology, fibrosis and congenital diseases. The knowledge about the mechanisms involved in TGF-β signal transduction has allowed a better understanding of the disease pathogenicity as well as the identification of several molecular targets with great potential in therapeutic interventions.


Journal of Biological Chemistry | 2002

Extracellular and cytoplasmic domains of endoglin interact with the transforming growth factor-beta receptors I and II

Mercedes Guerrero-Esteo; Tilman Sanchez-Elsner; Ainhoa Letamendía; Carmelo Bernabeu

Endoglin is an auxiliary component of the transforming growth factor-β (TGF-β) receptor system, able to associate with the signaling receptor types I (TβRI) and II (TβRII) in the presence of ligand and to modulate the cellular responses to TGF-β1. Endoglin cannot bind ligand on its own but requires the presence of the signaling receptors, supporting a critical role for the interaction between endoglin and TβRI or TβRII. This study shows that full-length endoglin interacts with both TβRI and TβRII, independently of their kinase activation state or the presence of exogenous TGF-β1. Truncated constructs encoding either the extracellular or the cytoplasmic domains of endoglin demonstrated that the association with the signaling receptors occurs through both extracellular and cytoplasmic domains. However, a more specific mapping revealed that the endoglin/TβRI interaction was different from that of endoglin/TβRII. TβRII interacts with the amino acid region 437–558 of the extracellular domain of endoglin, whereas TβRI interacts not only with the region 437–558 but also with the protein region located between amino acid 437 and the N terminus. Both TβRI and TβRII interact with the cytoplasmic domain of endoglin, but TβRI only interacts when the kinase domain is inactive, whereas TβRII remains associated in its active and inactive forms. Upon association, TβRI and TβRII phosphorylate the endoglin cytoplasmic domain, and then TβRI, but not TβRII, kinase dissociates from the complex. Conversely, endoglin expression results in an altered phosphorylation state of TβRII, TβRI, and downstream Smad proteins as well as a modulation of TGF-β signaling, as measured by the reporter gene expression. These results suggest that by interacting through its extracellular and cytoplasmic domains with the signaling receptors, endoglin might affect TGF-β responses.


Journal of Cellular Physiology | 2005

Interaction and functional interplay between endoglin and ALK‐1, two components of the endothelial transforming growth factor‐β receptor complex

Francisco J. Blanco; Juan Francisco Santibanez; Mercedes Guerrero-Esteo; Carmen Langa; Calvin P.H. Vary; Carmelo Bernabeu

Transforming growth factor‐β (TGF‐β) signaling in endothelial cells is able to modulate angiogenesis and vascular remodeling, although the underlying molecular mechanisms remain poorly understood. Endoglin and ALK‐1 are components of the TGF‐β receptor complex, predominantly expressed in endothelial cells, and mutations in either endoglin or ALK‐1 genes are responsible for the vascular dysplasia known as hereditary hemorrhagic telangiectasia. Here we find that the extracellular and cytoplasmic domains of the auxiliary TGF‐β receptor endoglin interact with ALK‐1 (a type I TGF‐β receptor). In addition, endoglin potentiates TGF‐β/ALK1 signaling, with the extracellular domain of endoglin contributing to this functional cooperation between endoglin and ALK‐1. By contrast, endoglin appears to interfere with TGF‐β/ALK‐5 signaling. These results suggest that the functional association of endoglin with ALK‐1 is critical for the endothelial responses to TGF‐β.


Blood | 2011

Activin A skews macrophage polarization by promoting a proinflammatory phenotype and inhibiting the acquisition of anti-inflammatory macrophage markers

Elena Sierra-Filardi; Amaya Puig-Kröger; Francisco J. Blanco; Concha Nieto; Rafael Bragado; M. Isabel Palomero; Carmelo Bernabeu; Miguel A. Vega; Angel L. Corbí

M-CSF favors the generation of folate receptor β-positive (FRβ⁺), IL-10-producing, immunosuppressive, M2-polarized macrophages [M2 (M-CSF)], whereas GM-CSF promotes a proinflammatory, M1-polarized phenotype [M1 (GM-CSF)]. In the present study, we found that activin A was preferentially released by M1 (GM-CSF) macrophages, impaired the acquisition of FRβ and other M2 (M-CSF)-specific markers, down-modulated the LPS-induced release of IL-10, and mediated the tumor cell growth-inhibitory activity of M1 (GM-CSF) macrophages, in which Smad2/3 is constitutively phosphorylated. The contribution of activin A to M1 (GM-CSF) macrophage polarization was evidenced by the capacity of a blocking anti-activin A antibody to reduce M1 (GM-CSF) polarization markers expression while enhancing FRβ and other M2 (M-CSF) markers mRNA levels. Moreover, an inhibitor of activin receptor-like kinase 4/5/7 (ALK4/5/7 or SB431542) promoted M2 (M-CSF) marker expression but limited the acquisition of M1 (GM-CSF) polarization markers, suggesting a role for Smad2/3 activation in macrophage polarization. In agreement with these results, expression of activin A and M2 (M-CSF)-specific markers was oppositely regulated by tumor ascites. Therefore, activin A contributes to the proinflammatory macrophage polarization triggered by GM-CSF and limits the acquisition of the anti-inflammatory phenotype in a Smad2-dependent manner. Our results demonstrate that activin A-initiated Smad signaling skews macrophage polarization toward the acquisition of a proinflammatory phenotype.


Journal of Cell Science | 2003

CD105 prevents apoptosis in hypoxic endothelial cells

Chenggang Li; Razao Issa; Patricia Kumar; Ian N. Hampson; José M. López-Novoa; Carmelo Bernabeu; Shant Kumar

CD105, a marker of endothelial cells, is abundantly expressed in tissues undergoing angiogenesis and is a receptor for transforming growth factorβ. The pivotal role of CD105 in the vascular system was demonstrated by the severe vascular defects that occur in CD105-knockout mice, but the exact mechanisms for CD105 regulation of vascular development have not been fully elucidated. In light of the function of CD105 and the importance of hypoxia in neovascularisation, we speculated that CD105 is involved in hypoxia-initiated angiogenesis. Using tissue-cultured human microvascular endothelial cells, we have investigated the effects of hypoxic stress on CD105 gene expression. Hypoxia induced a significant increase in membrane-bound and secreted CD105 protein levels. CD105 mRNA and promoter activity were also markedly elevated, the latter returning to the basal level after 16 hours of hypoxic stress. Hypoxia induced cell cycle arrest at the G0/G1 phases and massive cell apoptosis after 24 hours through a reduction in the Bcl-2 to Bax ratio, downregulation of Bcl-XL and Mcl-1, and upregulation of caspase-3 and caspase-8. The consequence of CD105 upregulation was revealed using an antisense approach and a TUNEL assay. Suppression of CD105 increased cell apoptosis under hypoxic stress in the absence of TGFβ1. Furthermore, hypoxia and TGFβ1 synergistically induced apoptosis in the CD105-deficient cells but not in the control cells. We conclude that hypoxia is a potent stimulus for CD105 gene expression in vascular endothelial cells, which in turn attenuates cell apoptosis and thus contributes to angiogenesis.


The FASEB Journal | 2004

Endoglin regulates nitric oxide-dependent vasodilatation

Mirjana Jerkic; Juan V. Rivas-Elena; Marta Prieto; Rosalía Carrón; Francisco Sanz-Rodríguez; Fernando Pérez-Barriocanal; Alicia Rodríguez-Barbero; Carmelo Bernabeu; José M. López-Novoa

Endoglin is a membrane glycoprotein that plays an important role in cardiovascular development and angiogenesis. We examined the role of endoglin in the control of vascular tone by measuring nitric oxide (NO)‐dependent vasodilation in haploinsufficient mice (Eng+/−) and their Eng+/+ littermates. The vasodilatory effect of acetylcholine, bradykinin, and sodium nitroprusside was assessed in anesthetized mice; in isolated, perfused hindlimbs; and in aortic rings. The substantial hypotensive and vasodilatory response induced by acetylcholine and bradykinin in Eng+/+ was markedly reduced in Eng+/− mice. Both kinds of animals had similar responses to sodium nitroprusside, suggesting that the deficient vasodilatory effect is not due to a NO response impairment. Urinary and plasma concentrations of nitrites, a NO metabolite, were lower in Eng+/− than in Eng+/+ mice. The levels of endothelial nitric oxide synthase (eNOS) in kidneys and femoral arteries were about half in Eng+/− than in Eng+/+ mice and were also reduced in primary cultures of aortic endothelial cells from Eng+/− compared with those from Eng+/+ mice. Furthermore, overexpression or suppression of endoglin in cultured cells induced a marked increase or decrease in the protein levels of eNOS, respectively. Thus, our results in vivo and in vitro demonstrate a relationship between endoglin and NO‐dependent vasodilation mediated by the regulation of eNOS expression.


American Journal of Physiology-heart and Circulatory Physiology | 2010

The physiological role of endoglin in the cardiovascular system

José M. López-Novoa; Carmelo Bernabeu

Endoglin (CD105) is an integral membrane glycoprotein that serves as a coreceptor for members of the transforming growth factor-β superfamily of proteins. A major role for endoglin in regulating transforming growth factor-β-dependent vascular remodeling and angiogenesis has been postulated based on the following: 1) endoglin is the gene mutated in hereditary hemorrhagic telangiectasia type 1, a disease characterized by vascular malformations; 2) endoglin knockout mice die at midgestation because of defective angiogenesis; 3) endoglin is overexpressed in neoangiogenic vessels, during inflammation, and in solid tumors; and 4) endoglin regulates the expression and activity of endothelial nitric oxide synthase, which is involved in angiogenesis and vascular tone. Besides the predominant form of the endoglin receptor (long endoglin isoform), two additional forms of endoglin have been recently reported to play a role in the vascular pathology and homeostasis: the alternatively spliced short endoglin isoform and a soluble endoglin form that is proteolytically cleaved from membrane-bound endoglin. The purpose of this review is to underline the role that the different forms of endoglin play in regulating angiogenesis, vascular remodeling, and vascular tone, as well as to analyze the molecular and cellular mechanisms supporting these effects.


Journal of Cellular Biochemistry | 2007

Novel biochemical pathways of endoglin in vascular cell physiology

Carmelo Bernabeu; Barbara A. Conley; Calvin P.H. Vary

The broad role of the transforming growth factor beta (TGFβ) signaling pathway in vascular development, homeostasis, and repair is well appreciated. Endoglin is emerging as a novel, complex, and poorly understood regulatory component of the TGFβ receptor complex, whose importance is underscored by its recognition as the site of mutations causing hereditary hemorrhagic telangiectasia (HHT) [McAllister et al., 1994 ]. Extensive analyses of endoglin function in normal developmental mouse models [Bourdeau et al., 1999 ; Li et al., 1999 ; Arthur et al., 2000 ] and in HHT animal models [Bourdeau et al., 2000 ; Torsney et al., 2003 ] exemplify the importance of understanding endoglins biochemical functions. However, novel mechanisms underlying the regulation of these pathways continue to emerge. These mechanisms include modification of TGFβ receptor signaling at the ligand and receptor activation level, direct effects of endoglin on cell adhesion and migration, and emerging roles for endoglin in the determination of stem cell fate and tissue patterning. The purpose of this review is to highlight the cellular and molecular studies that underscore the central role of endoglin in vascular development and disease. J. Cell. Biochem. 102: 1375–1388, 2007.


Atherosclerosis | 2000

Endoglin, a TGF-beta receptor-associated protein, is expressed by smooth muscle cells in human atherosclerotic plaques

Barbara A. Conley; Joshua D. Smith; Mercedes Guerrero-Esteo; Carmelo Bernabeu; Calvin P.H. Vary

Endoglin is a transmembrane protein that is found in association with transforming growth factor-beta (TGF-beta) superfamily receptor complexes and has an expression pattern that appears to be restricted primarily to endothelial cells, activated macrophages, trophoblasts, and fibroblasts. Since mutations in endoglin have been shown to be linked to hereditary hemorrhagic telangiectasia type 1, a disease manifested as vascular malformations characterized by excessive layers of vascular smooth muscle cells (VSMC), the expression of endoglin was investigated in VSMC. In vivo, the majority of SMC in human atherosclerotic plaques expressed high levels of endoglin, while endoglin was not detected in SMC from samples of the normal arterial wall. In vitro studies demonstrate that human aortic smooth muscle cells (HASMC) express the L-isoform of endoglin. Like endothelial cells, HASMC express endoglin protein as a dimer on the cell surface that binds TGF-beta1. In vitro, endoglin expression by HASMC is upregulated in response to TGF-beta1, suggesting that the presence of this factor in the atherosclerotic plaque might be responsible for the increased expression of endoglin. The demonstration of increased levels of endoglin in VSMC in human atherosclerotic plaques suggests a role for SMC endoglin in the maintenance of vascular integrity and in the response of the vessel wall to injury.


Blood | 2010

SETBP1 overexpression is a novel leukemogenic mechanism that predicts adverse outcome in elderly patients with acute myeloid leukemia

Ion Cristóbal; Francisco J. Blanco; Laura Garcia-Orti; Nerea Marcotegui; Carmen Vicente; José Rifón; Francisco J. Novo; Eva Bandrés; María José Calasanz; Carmelo Bernabeu; María D. Odero

Acute myeloid leukemias (AMLs) result from multiple genetic alterations in hematopoietic stem cells. We describe a novel t(12;18)(p13;q12) involving ETV6 in a patient with AML. The translocation resulted in overexpression of SETBP1 (18q12), located close to the breakpoint. Overexpression of SETBP1 through retroviral insertion has been reported to confer growth advantage in hematopoietic progenitor cells. We show that SETBP1 overexpression protects SET from protease cleavage, increasing the amount of full-length SET protein and leading to the formation of a SETBP1-SET-PP2A complex that results in PP2A inhibition, promoting proliferation of the leukemic cells. The prevalence of SETBP1 overexpression in AML at diagnosis (n = 192) was 27.6% and was associated with unfavorable cytogenetic prognostic group, monosomy 7, and EVI1 overexpression (P < .01). Patients with SETBP1 overexpression had a significantly shorter overall survival, and the prognosis impact was remarkably poor in patients older than 60 years in both overall survival (P = .015) and event-free survival (P = .015). In summary, our data show a novel leukemogenic mechanism through SETBP1 overexpression; moreover, multivariate analysis confirms the negative prognostic impact of SETBP1 overexpression in AML, especially in elderly patients, where it could be used as a predictive factor in any future clinical trials with PP2A activators.

Collaboration


Dive into the Carmelo Bernabeu's collaboration.

Top Co-Authors

Avatar

José M. López-Novoa

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Luisa María Botella

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Carmen Langa

University of Salamanca

View shared research outputs
Top Co-Authors

Avatar

Francisco J. Blanco

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Francisco Sanz-Rodríguez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miguel Arévalo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miguel Pericacho

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge