Network


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

Hotspot


Dive into the research topics where Valentín Mendoza is active.

Publication


Featured researches published by Valentín Mendoza.


Biochemical Journal | 2001

Recombinant soluble betaglycan is a potent and isoform-selective transforming growth factor-beta neutralizing agent.

M. Magdalena Vilchis-Landeros; Jose L. Montiel; Valentín Mendoza; Guillermo Mendoza-Hernández; Fernando López-Casillas

Betaglycan is an accessory receptor of members of the transforming growth factor-beta (TGF-beta) superfamily, which regulates their actions through ligand-dependent interactions with type II receptors. A natural soluble form of betaglycan is found in serum and extracellular matrices. Soluble betaglycan, prepared as a recombinant protein using the baculoviral expression system, inhibits the actions of TGF-beta. Because of its potential use as an anti-TGF-beta therapeutic agent, we have purified and characterized baculoviral recombinant soluble betaglycan. Baculoviral soluble betaglycan is a homodimer formed by two 110 kDa monomers associated by non-covalent interactions. This protein is devoid of glycosaminoglycan chains, although it contains the serine residues, which, in vertebrate cells, are modified by these carbohydrates. On the other hand, mannose-rich carbohydrates account for approximately 20 kDa of the mass of the monomer. End-terminal sequence analysis of the soluble betaglycan showed that Gly(24) is the first residue of the mature protein. Similarly to the natural soluble betaglycan, baculoviral soluble betaglycan has an equilibrium dissociation constant (K(d)) of 3.5 nM for TGF-beta1. Ligand competition assays indicate that the relative affinities of recombinant soluble betaglycan for the TGF-beta isoforms are TGF-beta2>TGF-beta3>TGF-beta1. The anti-TGF-beta potency of recombinant soluble betaglycan in vitro is 10-fold higher for TGF-beta2 than for TGF-beta1. Compared with a commercial pan-specific anti-TGF-beta neutralizing antibody, recombinant soluble betaglycan is more potent against TGF-beta2 and similar against TGF-beta1. These results indicate that baculoviral soluble betaglycan has the biochemical and functional properties that would make it a suitable agent for the treatment of the diseases in which excess TGF-beta plays a central physiopathological role.


Journal of Biological Chemistry | 2002

p32 (gC1qBP) Is a General Protein Kinase C (PKC)-binding Protein INTERACTION AND CELLULAR LOCALIZATION OF P32-PKC COMPLEXES IN RAT HEPATOCYTES

Martha Robles-Flores; Erika Rendón-Huerta; Héctor González-Aguilar; Guillermo Mendoza-Hernández; Socorro Islas; Valentín Mendoza; M. Verónica Ponce-Castañeda; Lorenza González-Mariscal; Fernando López-Casillas

The aim of this study was to identify cellular proteins that bind protein kinase C (PKC) and may influence its activity and its localization. A 32-kDa PKC-binding protein was purified to homogeneity from the Triton X-100-insoluble fraction obtained from hepatocytes homogenates. The protein was identified by NH2-terminal amino acid sequencing as the previously described mature form of p32 (gC1qR). Recombinant p32 was expressed as a glutathione S-transferase fusion protein, affinity-purified, and tested for an in vitro interaction with PKC using an overlay assay approach. All PKC isoforms expressed in rat hepatocytes interacted in vitro with p32, but the binding dependence on PKC activators was different for each one. Whereas PKCδ only binds to p32 in the presence of PKC activators, PKCζ and PKCα increase their binding when they are in the activated form. Other PKC isoforms such as β, ε, and θ bind equally well to p32 regardless of the presence of PKC activators, and PKCμ binds even better in their absence. It was also found that p32 is not a substrate for any of the PKC isoforms tested, but interestingly, its presence had a stimulatory effect (2-fold for PKCδ) on PKC activity. We also observed in vivo interaction between PKC and p32 by immunofluorescence and confocal microscopy. A time course of phorbol ester treatment of cultured rat hepatocytes (C9 cells) showed that PKCθ and p32 are constitutively associated in vivo, whereas PKCδ activation is required for its association with p32. Our data also showed that phorbol ester treatment induces a transient translocation of p32 from the cytoplasm to the cell nucleus. Together, these findings suggest that p32 may be a regulator of PKC location and function.


Journal of Clinical Investigation | 2008

Vasoinhibins prevent retinal vasopermeability associated with diabetic retinopathy in rats via protein phosphatase 2A–dependent eNOS inactivation

Celina García; Jorge Aranda; Edith Arnold; Stéphanie Thebault; Yazmín Macotela; Fernando López-Casillas; Valentín Mendoza; Hugo Quiroz-Mercado; Hebert Luis Hernández-Montiel; Sue Hwa Lin; Gonzalo Martínez de la Escalera; Carmen Clapp

Increased retinal vasopermeability contributes to diabetic retinopathy, the leading cause of blindness in working-age adults. Despite clinical progress, effective therapy remains a major need. Vasoinhibins, a family of peptides derived from the protein hormone prolactin (and inclusive of the 16-kDa fragment of prolactin), antagonize the proangiogenic effects of VEGF, a primary mediator of retinal vasopermeability. Here, we demonstrate what we believe to be a novel function of vasoinhibins as inhibitors of the increased retinal vasopermeability associated with diabetic retinopathy. Vasoinhibins inhibited VEGF-induced vasopermeability in bovine aortic and rat retinal capillary endothelial cells in vitro. In vivo, vasoinhibins blocked retinal vasopermeability in diabetic rats and in response to intravitreous injection of VEGF or of vitreous from patients with diabetic retinopathy. Inhibition by vasoinhibins was similar to that achieved following immunodepletion of VEGF from human diabetic retinopathy vitreous or blockage of NO synthesis, suggesting that vasoinhibins inhibit VEGF-induced NOS activation. We further showed that vasoinhibins activate protein phosphatase 2A (PP2A), leading to eNOS dephosphorylation at Ser1179 and, thereby, eNOS inactivation. Moreover, intravitreous injection of okadaic acid, a PP2A inhibitor, blocked the vasoinhibin effect on endothelial cell permeability and retinal vasopermeability. These results suggest that vasoinhibins have the potential to be developed as new therapeutic agents to control the excessive retinal vasopermeability observed in diabetic retinopathy and other vasoproliferative retinopathies.


The EMBO Journal | 2011

TGF-β signalling is mediated by two autonomously functioning TβRI:TβRII pairs

Tao Huang; Laurent David; Valentín Mendoza; Yong Yang; Maria M. Villarreal; Keya De; Lu-Zhe Sun; Xiaohong Fang; Fernando López-Casillas; Jeffrey L. Wrana; Andrew P. Hinck

Transforming growth factor (TGF)‐βs are dimeric polypeptides that have vital roles in regulating cell growth and differentiation. They signal by assembling a receptor heterotetramer composed of two TβRI:TβRII heterodimers. To investigate whether the two heterodimers bind and signal autonomously, one of the TGF‐β protomers was substituted to block receptor binding. The substituted dimer, TGF‐β3 WD, bound the TβRII extracellular domain and recruited the TβRI with affinities indistinguishable from TGF‐β3, but with one‐half the stoichiometry. TGF‐β3 WD was further shown to retain one‐quarter to one‐half the signalling activity of TGF‐β3 in three established assays for TGF‐β function. Single‐molecule fluorescence imaging with GFP‐tagged receptors demonstrated a measurable increase in the proportion of TβRI and TβRII dimers upon treatment with TGF‐β3, but not with TGF‐β3 WD. These results provide evidence that the two TβRI:TβRII heterodimers bind and signal in an autonomous manner. They further underscore how the TGF‐βs diverged from the bone morphogenetic proteins, the ancestral ligands of the TGF‐β superfamily that signal through a RI:RII:RII heterotrimer.


Biochemistry | 2009

Betaglycan has Two Independent Domains Required for High Affinity TGF-β Binding: Proteolytic Cleavage Separates the Domains and Inactivates the Neutralizing Activity of the Soluble Receptor

Valentín Mendoza; M. Magdalena Vilchis-Landeros; Guillermo Mendoza-Hernández; Tao Huang; Maria M. Villarreal; Andrew P. Hinck; Fernando López-Casillas; José Luis Montiel

Betaglycan is a coreceptor for members of the transforming growth factor beta (TGF-beta) superfamily. Mutagenesis has identified two ligand binding regions, one at the membrane-distal and the other at the membrane-proximal half of the betaglycan ectodomain. Here we show that partial plasmin digestion of soluble betaglycan produces two proteolysis-resistant fragments of 45 and 55 kDa, consistent with the predicted secondary structure, which indicates an intervening nonstructured linker region separating the highly structured N- and C-terminal domains. Amino terminal sequencing indicates that the 45 and 55 kDa fragments correspond, respectively, to the membrane-distal and -proximal regions. Plasmin treatment of membrane betaglycan results in the production of equivalent proteolysis-resistant fragments. The 45 and 55 kDa fragments, as well as their recombinant soluble counterparts, Sol Delta10 and Sol Delta11, bind TGF-beta, but nonetheless, compared to intact soluble betaglycan, have a severely diminished ability to block TGF-beta activity. Surface plasmon resonance (SPR) analysis indicates that soluble betaglycan has K(d)s in the low nanomolar range for the three TGF-beta isoforms, while those for Sol Delta10 and Sol Delta11 are 1-2 orders of magnitude higher. SPR analysis further shows that the K(d)s of Sol Delta11 are not changed in the presence of Sol Delta10, indicating that the high affinity of soluble betaglycan is a consequence of tethering the domains together. Overall, these results suggest that betaglycan ectodomain exhibits a bilobular structure in which each lobule folds independently and binds TGF-beta through distinct nonoverlapping interfaces and that linker modification may be an approach to improve soluble betaglycans TGF-beta neutralizing activity.


Cellular and Molecular Life Sciences | 2015

Proteolytic control of TGF-β co-receptor activity by BMP-1/tolloid-like proteases revealed by quantitative iTRAQ proteomics.

Frédéric Delolme; Cyril Anastasi; Lindsay B. Alcaraz; Valentín Mendoza; Sandrine Vadon-Le Goff; Maya Talantikite; Robin Capomaccio; Jimmy Mevaere; Laëtitia Fortin; Dominique Mazzocut; Odile Damour; Isabelle Zanella-Cléon; David J. S. Hulmes; Christopher M. Overall; Ulrich Valcourt; Fernando López-Casillas; Catherine Moali

The metalloproteinase BMP-1 (bone morphogenetic protein-1) plays a major role in the control of extracellular matrix (ECM) assembly and growth factor activation. Most of the growth factors activated by BMP-1 are members of the TGF-β superfamily known to regulate multiple biological processes including embryonic development, wound healing, inflammation and tumor progression. In this study, we used an iTRAQ (isobaric tags for relative and absolute quantification)-based quantitative proteomic approach to reveal the release of proteolytic fragments from the cell surface or the ECM by BMP-1. Thirty-eight extracellular proteins were found in significantly higher or lower amounts in the conditioned medium of HT1080 cells overexpressing BMP-1 and thus, could be considered as candidate substrates. Strikingly, three of these new candidates (betaglycan, CD109 and neuropilin-1) were TGF-β co-receptors, also acting as antagonists when released from the cell surface, and were chosen for further substrate validation. Betaglycan and CD109 proved to be directly cleaved by BMP-1 and the corresponding cleavage sites were extensively characterized using a new mass spectrometry approach. Furthermore, we could show that the ability of betaglycan and CD109 to interact with TGF-β was altered after cleavage by BMP-1, leading to increased and prolonged SMAD2 phosphorylation in BMP-1-overexpressing cells. Betaglycan processing was also observed in primary corneal keratocytes, indicating a general and novel mechanism by which BMP-1 directly affects signaling by controlling TGF-β co-receptor activity. The proteomic data have been submitted to ProteomeXchange with the identifier PXD000786 and doi:10.6019/PXD000786.


Biochemistry | 2016

Binding Properties of the Transforming Growth Factor-β Coreceptor Betaglycan: Proposed Mechanism for Potentiation of Receptor Complex Assembly and Signaling

Maria M. Villarreal; Sun Kyung Kim; Lindsey Barron; Ravi Kodali; Jason Baardsnes; Cynthia S. Hinck; Troy C. Krzysiak; Morkos A. Henen; Olga N. Pakhomova; Valentín Mendoza; Maureen O’Connor-McCourt; Eileen M. Lafer; Fernando López-Casillas; Andrew P. Hinck

Transforming growth factor (TGF) β1, β2, and β3 (TGF-β1–TGF-β3, respectively) are small secreted signaling proteins that each signal through the TGF-β type I and type II receptors (TβRI and TβRII, respectively). However, TGF-β2, which is well-known to bind TβRII several hundred-fold more weakly than TGF-β1 and TGF-β3, has an additional requirement for betaglycan, a membrane-anchored nonsignaling receptor. Betaglycan has two domains that bind TGF-β2 at independent sites, but how it binds TGF-β2 to potentiate TβRII binding and how the complex with TGF-β, TβRII, and betaglycan undergoes the transition to the signaling complex with TGF-β, TβRII, and TβRI are not understood. To investigate the mechanism, the binding of the TGF-βs to the betaglycan extracellular domain, as well as its two independent binding domains, either directly or in combination with the TβRI and TβRII ectodomains, was studied using surface plasmon resonance, isothermal titration calorimetry, and size-exclusion chromatography. These studies show that betaglycan binds TGF-β homodimers with a 1:1 stoichiometry in a manner that allows one molecule of TβRII to bind. These studies further show that betaglycan modestly potentiates the binding of TβRII and must be displaced to allow TβRI to bind. These findings suggest that betaglycan functions to bind and concentrate TGF-β2 on the cell surface and thus promote the binding of TβRII by both membrane-localization effects and allostery. These studies further suggest that the transition to the signaling complex is mediated by the recruitment of TβRI, which simultaneously displaces betaglycan and stabilizes the bound TβRII by direct receptor–receptor contact.


Current Genetics | 1987

Extrachromosomal genetics in the yeast Kluyveromyces lactis: isolation and characterization of antimycin-resistant mutants

Aurora L. Brummer; Valentín Mendoza; Alba Tuena de Cobo

SummaryAntimycin-resistant (AR) mutants of the yeast Kluyveromyces lactis, obtained either spontaneously or after manganese treatment, were isolated and genetically characterized. Most of the mutants obtained after manganese mutagenesis and two spontaneous mutants, tolerated high antimycin concentrations (more than 10 /gmg/ml) and were extrachromosomal. One mutant which grew only in low antimycin (1 /gmg/ml) showed a Mendelian type of inheritance. The extrachromosomal mutants could be assigned to at least two genetic loci (AIRand AIIR). Mutants representative of these two groups showed increased resistance to the antibiotic when the respiration of whole cells or mitochondria was studied. Extrachromosomal mutants of Saccharomyces cerevisiae resistant to antimycin were also induced with manganese, isolated and characterized. Comparative studies of the antimycin-resistant mutants of K. lactis and S. cerevisiae permitted the following observations: a) K. lactis is more resistant to antimycin, funiculosin, mucidin and diuron than S. cerevisiae, as are the AR mutants; b) K. lactis shows correlated sensitivity to funiculosin differing in this aspect from S. cerevisiae; c) the antimycin-resistant mutants of K. lactis belonging to group 11 (AIIR) were also resistant to diuron, tolerating concentrations of more than 200 /gmg/ml; d) all extrachromosomal antimycin-resistant-mutants of S. cerevisiae and some of the AR mutants of K. lactis were more sensitive to mucidin than the wild type.


Genesis | 2015

Betaglycan knock‐down causes embryonic angiogenesis defects in zebrafish

Andrés Kamaid; Tonatiuh Molina-Villa; Valentín Mendoza; Cristina Pujades; Ernesto Maldonado; Juan Carlos Ispizua Belmonte; Fernando López-Casillas

Angiogenesis is an essential requirement for embryonic development and adult homeostasis. Its deregulation is a key feature of numerous pathologies and many studies have shown that members of the transforming growth factor beta (TGF‐β) family of proteins play important roles in angiogenesis during development and disease. Betaglycan (BG), also known as TGF‐β receptor type III, is a TGF‐β coreceptor essential for mice embryonic development but its role in angiogenesis has not been described. We have cloned the cDNA encoding zebrafish BG, a TGF‐β‐binding membrane proteoglycan that showed a dynamic expression pattern in zebrafish embryos, including the notochord and cells adjacent to developing vessels. Injection of antisense morpholinos decreased BG protein levels and morphant embryos exhibited impaired angiogenesis that was rescued by coinjection with rat BG mRNA. In vivo time‐lapse microscopy revealed that BG deficiency differentially affected arterial and venous angiogenesis: morphants showed impaired pathfinding of intersegmental vessels migrating from dorsal aorta, while endothelial cells originating from the caudal vein displayed sprouting and migration defects. Our results reveal a new role for BG during embryonic angiogenesis in zebrafish, which has not been described in mammals and pose interesting questions about the molecular machinery regulating angiogenesis in different vertebrates. genesis 53:583–603, 2015.


PLOS ONE | 2012

Betaglycan (TβRIII) is expressed in the thymus and regulates T cell development by protecting thymocytes from apoptosis.

Germán R. Alemán-Muench; Valentín Mendoza; Kaye L. Stenvers; Eduardo A. García-Zepeda; Fernando López-Casillas; Chander Raman; Gloria Soldevila

TGF-β type III receptor (TβRIII) is a coreceptor for TGFβ family members required for high-affinity binding of these ligands to their receptors, potentiating their cellular functions. TGF-β [1]–[3], bone morphogenetic proteins (BMP2/4) and inhibins regulate different checkpoints during T cell differentiation. Although TβRIII is expressed on hematopoietic cells, the role of this receptor in the immune system remains elusive. Here, we provide the first evidence that TβRIII is developmentally expressed during T cell ontogeny, and plays a crucial role in thymocyte differentiation. Blocking of endogenous TβRIII in fetal thymic organ cultures led to a delay in DN-DP transition. In addition, in vitro development of TβRIII−/− thymic lobes also showed a significant reduction in absolute thymocyte numbers, which correlated with increased thymocyte apoptosis, resembling the phenotype reported in Inhibin α −/− thymic lobes. These data suggest that Inhibins and TβRIII may function as a molecular pair regulating T cell development.

Collaboration


Dive into the Valentín Mendoza's collaboration.

Top Co-Authors

Avatar

Fernando López-Casillas

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

LuZhe Sun

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Cynthia S. Hinck

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Junhua Yang

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

M. Magdalena Vilchis-Landeros

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Jorge Zúñiga

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Maria M. Villarreal

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Yuping Tang

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Guillermo Mendoza-Hernández

National Autonomous University of Mexico

View shared research outputs
Researchain Logo
Decentralizing Knowledge