Network


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

Hotspot


Dive into the research topics where Jose L. Izquierdo-Garcia is active.

Publication


Featured researches published by Jose L. Izquierdo-Garcia.


Nature Medicine | 2013

Mutations in the NOTCH pathway regulator MIB1 cause left ventricular noncompaction cardiomyopathy

Guillermo Luxán; Jesús C. Casanova; Beatriz Martínez-Poveda; Belén Prados; Gaetano D'Amato; Donal MacGrogan; Alvaro González-Rajal; David Dobarro; Carlos Torroja; Fernando J. Martinez; Jose L. Izquierdo-Garcia; Leticia Fernández-Friera; María Sabater-Molina; Young-Y Kong; Gonzalo Pizarro; Borja Ibanez; Constancio Medrano; Pablo García-Pavía; Juan R. Gimeno; Lorenzo Monserrat; Luis Jesús Jiménez-Borreguero; José Luis de la Pompa

Left ventricular noncompaction (LVNC) causes prominent ventricular trabeculations and reduces cardiac systolic function. The clinical presentation of LVNC ranges from asymptomatic to heart failure. We show that germline mutations in human MIB1 (mindbomb homolog 1), which encodes an E3 ubiquitin ligase that promotes endocytosis of the NOTCH ligands DELTA and JAGGED, cause LVNC in autosomal-dominant pedigrees, with affected individuals showing reduced NOTCH1 activity and reduced expression of target genes. Functional studies in cells and zebrafish embryos and in silico modeling indicate that MIB1 functions as a dimer, which is disrupted by the human mutations. Targeted inactivation of Mib1 in mouse myocardium causes LVNC, a phenotype mimicked by inactivation of myocardial Jagged1 or endocardial Notch1. Myocardial Mib1 mutants show reduced ventricular Notch1 activity, expansion of compact myocardium to proliferative, immature trabeculae and abnormal expression of cardiac development and disease genes. These results implicate NOTCH signaling in LVNC and indicate that MIB1 mutations arrest chamber myocardium development, preventing trabecular maturation and compaction.


Progress in Nuclear Magnetic Resonance Spectroscopy | 2011

Descriptive review of current NMR-based metabolomic data analysis packages

Jose L. Izquierdo-Garcia; Palmira Villa; Angelos Kyriazis; Laura del Puerto-Nevado; Sandra Pérez-Rial; Ignacio R. Rodriguez; Natalia Hernandez; Jesús Ruiz-Cabello

CIBERES, CIBER Enfermedades Respiratorias, Departartamento Quimica-Fisica II, Facultad Farmacia, Universidad Complutense de Madrid, Madrid, Spain CIBERES, CIBER Enfermedades Respiratorias, CAI de Resonancia Magnetica, Universidad Complutense de Madrid, Madrid, Spain CIBERES, CIBER Enfermedades Respiratorias, Fundacion Jimenez Diaz – CAPIO, Madrid, Spain Departamento de Ingenieria Rural, ETSI Agronomos, Universidad Politecnica de Madrid, Madrid, Spain


Cancer Research | 2015

IDH1 Mutation Induces Reprogramming of Pyruvate Metabolism.

Jose L. Izquierdo-Garcia; Pavithra Viswanath; Pia Eriksson; Larry Cai; Marina Radoul; Myriam M. Chaumeil; Michael D. Blough; H. Artee Luchman; Samuel Weiss; J. Gregory Cairncross; Joanna J. Phillips; Russell O. Pieper; Sabrina M. Ronen

Mutant isocitrate dehydrogenase 1 (IDH1) catalyzes the production of 2-hydroxyglutarate but also elicits additional metabolic changes. Levels of both glutamate and pyruvate dehydrogenase (PDH) activity have been shown to be affected in U87 glioblastoma cells or normal human astrocyte (NHA) cells expressing mutant IDH1, as compared with cells expressing wild-type IDH1. In this study, we show how these phenomena are linked through the effects of IDH1 mutation, which also reprograms pyruvate metabolism. Reduced PDH activity in U87 glioblastoma and NHA IDH1 mutant cells was associated with relative increases in PDH inhibitory phosphorylation, expression of pyruvate dehydrogenase kinase-3, and levels of hypoxia inducible factor-1α. PDH activity was monitored in these cells by hyperpolarized (13)C-magnetic resonance spectroscopy ((13)C-MRS), which revealed a reduction in metabolism of hyperpolarized 2-(13)C-pyruvate to 5-(13)C-glutamate, relative to cells expressing wild-type IDH1. (13)C-MRS also revealed a reduction in glucose flux to glutamate in IDH1 mutant cells. Notably, pharmacological activation of PDH by cell exposure to dichloroacetate (DCA) increased production of hyperpolarized 5-(13)C-glutamate in IDH1 mutant cells. Furthermore, DCA treatment also abrogated the clonogenic advantage conferred by IDH1 mutation. Using patient-derived mutant IDH1 neurosphere models, we showed that PDH activity was essential for cell proliferation. Taken together, our results established that the IDH1 mutation induces an MRS-detectable reprogramming of pyruvate metabolism, which is essential for cell proliferation and clonogenicity, with immediate therapeutic implications.


PLOS ONE | 2015

Metabolic Reprogramming in Mutant IDH1 Glioma Cells

Jose L. Izquierdo-Garcia; Pavithra Viswanath; Pia Eriksson; Myriam M. Chaumeil; Russell O. Pieper; Joanna J. Phillips; Sabrina M. Ronen

Background Mutations in isocitrate dehydrogenase (IDH) 1 have been reported in over 70% of low-grade gliomas and secondary glioblastomas. IDH1 is the enzyme that catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate while mutant IDH1 catalyzes the conversion of α-ketoglutarate into 2-hydroxyglutarate. These mutations are associated with the accumulation of 2-hydroxyglutarate within the tumor and are believed to be one of the earliest events in the development of low-grade gliomas. The goal of this work was to determine whether the IDH1 mutation leads to additional magnetic resonance spectroscopy (MRS)–detectable changes in the cellular metabolome. Methods Two genetically engineered cell models were investigated, a U87-based model and an E6/E7/hTERT immortalized normal human astrocyte (NHA)-based model. For both models, wild-type IDH1 cells were generated by transduction with a lentiviral vector coding for the wild-type IDH1 gene while mutant IDH1 cells were generated by transduction with a lentiviral vector coding for the R132H IDH1 mutant gene. Metabolites were extracted from the cells using the dual-phase extraction method and analyzed by 1H-MRS. Principal Component Analysis was used to analyze the MRS data. Results Principal Component Analysis clearly discriminated between wild-type and mutant IDH1 cells. Analysis of the loading plots revealed significant metabolic changes associated with the IDH1 mutation. Specifically, a significant drop in the concentration of glutamate, lactate and phosphocholine as well as the expected elevation in 2-hydroxyglutarate were observed in mutant IDH1 cells when compared to their wild-type counterparts. Conclusion The IDH1 mutation leads to several, potentially translatable MRS-detectable metabolic changes beyond the production of 2-hydroxyglutarate.


PLOS ONE | 2013

HDAC Inhibition Induces Increased Choline Uptake and Elevated Phosphocholine Levels in MCF7 Breast Cancer Cells

Christopher S. Ward; Pia Eriksson; Jose L. Izquierdo-Garcia; Alissa H. Brandes; Sabrina M. Ronen

Histone deacetylase (HDAC) inhibitors have emerged as effective antineoplastic agents in the clinic. Studies from our lab and others have reported that magnetic resonance spectroscopy (MRS)-detectable phosphocholine (PC) is elevated following SAHA treatment, providing a potential noninvasive biomarker of response. Typically, elevated PC is associated with cancer while a decrease in PC accompanies response to antineoplastic treatment. The goal of this study was therefore to elucidate the underlying biochemical mechanism by which HDAC inhibition leads to elevated PC. We investigated the effect of SAHA on MCF-7 breast cancer cells using 13C MRS to monitor [1,2-13C] choline uptake and phosphorylation to PC. We found that PC synthesis was significantly higher in treated cells, representing 154±19% of control. This was within standard deviation of the increase in total PC levels detected by 31P MRS (129±7% of control). Furthermore, cellular choline kinase activity was elevated (177±31%), while cytidylyltransferase activity was unchanged. Expression of the intermediate-affinity choline transporter SLC44A1 and choline kinase α increased (144% and 161%, respectively) relative to control, as determined by mRNA microarray analysis with protein-level confirmation by Western blotting. Taken together, our findings indicate that the increase in PC levels following SAHA treatment results from its elevated synthesis. Additionally, the concentration of glycerophosphocholine (GPC) increased significantly with treatment to 210±45%. This is likely due to the upregulated expression of several phospholipase A2 (PLA2) isoforms, resulting in increased PLA2 activity (162±18%) in SAHA-treated cells. Importantly, the levels of total choline (tCho)-containing metabolites, comprised of choline, PC and GPC, are readily detectable clinically using 1H MRS. Our findings thus provide an important step in validating clinically translatable non-invasive imaging methods for follow-up diagnostics of HDAC inhibitor treatment.


PLOS ONE | 2014

Glioma Cells with the IDH1 Mutation Modulate Metabolic Fractional Flux through Pyruvate Carboxylase

Jose L. Izquierdo-Garcia; Larry Cai; Myriam M. Chaumeil; Pia Eriksson; Aaron E. Robinson; Russell O. Pieper; Joanna J. Phillips; Sabrina M. Ronen

Background Over 70% of low-grade gliomas carry a heterozygous R132H mutation in the gene coding for isocitrate dehydrogenase 1 (IDH1). This confers the enzyme with the novel ability to convert α-ketoglutarate to 2-hydroxyglutarate, ultimately leading to tumorigenesis. The major source of 2-hydroxyglutarate production is glutamine, which, in cancer, is also a source for tricarboxylic acid cycle (TCA) anaplerosis. An alternate source of anaplerosis is pyruvate flux via pyruvate carboxylase (PC), which is a common pathway in normal astrocytes. The goal of this study was to determine whether PC serves as a source of TCA anaplerosis in IDH1 mutant cells wherein glutamine is used for 2-hydroxyglutarate production. Methods Immortalized normal human astrocytes engineered to express heterozygous mutant IDH1 or wild-type IDH1 were investigated. Flux of pyruvate via PC and via pyruvate dehydrogenase (PDH) was determined by using magnetic resonance spectroscopy to probe the labeling of [2-13C]glucose-derived 13C-labeled glutamate and glutamine. Activity assays, RT-PCR and western blotting were used to probe the expression and activity of relevant enzymes. The Cancer Genome Atlas (TCGA) data was analyzed to assess the expression of enzymes in human glioma samples. Results Compared to wild-type cells, mutant IDH1 cells significantly increased fractional flux through PC. This was associated with a significant increase in PC activity and expression. Concurrently, PDH activity significantly decreased, likely mediated by significantly increased inhibitory PDH phosphorylation by PDH kinase 3. Consistent with the observation in cells, analysis of TCGA data indicated a significant increase in PC expression in mutant IDH-expressing human glioma samples compared to wild-type IDH. Conclusions Our findings suggest that changes in PC and PDH may be an important part of cellular adaptation to the IDH1 mutation and may serve as potential therapeutic targets.


European Respiratory Journal | 2011

Is NMR-based metabolomic analysis of exhaled breath condensate accurate?

Jose L. Izquierdo-Garcia; Germán Peces-Barba; S. Heili; R. Diaz; E. Want; Jesús Ruiz-Cabello

To the Editors: The metabolomic analysis of exhaled breath condensate (EBC) is a simple noninvasive approach for the study of respiratory system diseases. Previous studies introduced nuclear magnetic resonance (NMR)-based metabolomics as a method allowing a definite separation between healthy patients and patients with airway disease 1, 2. In these studies, the influence of external contaminants was also considered. de Laurentiis et al. 1 reported that the removal of interfering residual external contaminants was crucial for correct EBC analysis and they proposed a cleaning protocol for the “complete removal of the disinfectant signals” from the reusable parts of the condensers. In order to verify the influence of the disinfectant signals we have compared the EBC 1H-NMR spectra of a healthy subject obtained after the standard cleaning protocol (disinfected for 15 min using a 1.5% Descogen™ solution and flushed for 15 min with water) recommended by the manufacturer’s guidelines and the International Consensus on EBC (fig. 1a) 3; one that was obtained after the cleaning protocol proposed by de Laurentiis et al. 1 (fig. 1b); and one obtained using a device without reusable condenser parts (fig. 1c). EBCs have been collected using an Anacon condenser (Biostec, Valencia, Spain). Sample collection and concentration (>99% water) were similar to those obtained in the procedure described by de Laurentiis …


Anesthesiology | 2014

A Metabolomic Approach to the Pathogenesis of Ventilator-induced Lung Injury.

Jose L. Izquierdo-Garcia; Shama Naz; Nicolás Nin; Yeny Rojas; Marcela Erazo; Leticia Martínez-Caro; Antonia García; Marta de Paula; Pilar Fernández-Segoviano; Cristina Casals; Andrés Esteban; Jesús Ruiz-Cabello; Coral Barbas; José A. Lorente

Background:Global metabolic profiling using quantitative nuclear magnetic resonance spectroscopy (MRS) and mass spectrometry (MS) is useful for biomarker discovery. The objective of this study was to discover biomarkers of acute lung injury induced by mechanical ventilation (ventilator-induced lung injury [VILI]), by using MRS and MS. Methods:Male Sprague–Dawley rats were subjected to two ventilatory strategies for 2.5 h: tidal volume 9 ml/kg, positive end-expiratory pressure 5 cm H2O (control, n = 14); and tidal volume 25 ml/kg and positive end-expiratory pressure 0 cm H2O (VILI, n = 10). Lung tissue, bronchoalveolar lavage fluid, and serum spectra were obtained by high-resolution magic angle spinning and 1H-MRS. Serum spectra were acquired by high-performance liquid chromatography coupled to quadupole-time of flight MS. Principal component and partial least squares analyses were performed. Results:Metabolic profiling discriminated characteristics between control and VILI animals. As compared with the controls, animals with VILI showed by MRS higher concentrations of lactate and lower concentration of glucose and glycine in lung tissue, accompanied by increased levels of glucose, lactate, acetate, 3-hydroxybutyrate, and creatine in bronchoalveolar lavage fluid. In serum, increased levels of phosphatidylcholine, oleamide, sphinganine, hexadecenal and lysine, and decreased levels of lyso-phosphatidylcholine and sphingosine were identified by MS. Conclusions:This pilot study suggests that VILI is characterized by a particular metabolic profile that can be identified by MRS and MS. The metabolic profile, though preliminary and pending confirmation in larger data sets, suggests alterations in energy and membrane lipids.SUPPLEMENTAL DIGITAL CONTENT IS AVAILABLE IN THE TEXT


PLOS ONE | 2016

New Biochemical Insights into the Mechanisms of Pulmonary Arterial Hypertension in Humans.

Renata Bujak; Jesús Mateo; Isabel Blanco; Jose L. Izquierdo-Garcia; Danuta Dudzik; Michał J. Markuszewski; Victor I. Peinado; Martin Laclaustra; Joan Albert Barberà; Coral Barbas; Jesús Ruiz-Cabello

Diagnosis of pulmonary arterial hypertension (PAH) is difficult due to the lack of specific clinical symptoms and biomarkers, especially at early stages. We compared plasma metabolic fingerprints of PAH patients (n = 20) with matched healthy volunteers (n = 20) using, for the first time, untargeted multiplatform metabolomics approach consisting of high-performance liquid and gas chromatography coupled with mass spectrometry. Multivariate statistical analyses were performed to select metabolites that contribute most to groups’ classification (21 from liquid in both ionization modes and 9 from gas chromatography-mass spectrometry). We found metabolites related to energy imbalance, such as glycolysis-derived metabolites, as well as metabolites involved in fatty acid, lipid and amino acid metabolism. We observed statistically significant changes in threitol and aminomalonic acid in PAH patients, which could provide new biochemical insights into the pathogenesis of the disease. The results were externally validated on independent case and control cohorts, confirming up to 16 metabolites as statistically significant in the validation study. Multiplatform metabolomics, followed by multivariate chemometric data analysis has a huge potential for explaining pathogenesis of PAH and for searching potential and new more specific and less invasive markers of the disease.


Oncotarget | 2016

Mutant IDH1 expression is associated with down-regulation of monocarboxylate transporters

Pavithra Viswanath; Chloe Najac; Jose L. Izquierdo-Garcia; Aleksandr Pankov; Chibo Hong; Pia Eriksson; Joseph F. Costello; Russell O. Pieper; Sabrina M. Ronen

Mutations in isocitrate dehydrogenase 1 (IDH1) are characteristic of low-grade gliomas. We recently showed that mutant IDH1 cells reprogram cellular metabolism by down-regulating pyruvate dehydrogenase (PDH) activity. Reduced pyruvate metabolism via PDH could lead to increased pyruvate conversion to lactate. The goal of this study was therefore to investigate the impact of the IDH1 mutation on the pyruvate-to-lactate flux. We used 13C magnetic resonance spectroscopy and compared the conversion of hyperpolarized [1-13C]-pyruvate to [1-13C]-lactate in immortalized normal human astrocytes expressing mutant or wild-type IDH1 (NHAIDHmut and NHAIDHwt). Our results indicate that hyperpolarized lactate production is reduced in NHAIDHmut cells compared to NHAIDHwt. This reduction was associated with lower expression of the monocarboxylate transporters MCT1 and MCT4 in NHAIDHmut cells. Furthermore, hyperpolarized lactate production was comparable in lysates of NHAIDHmut and NHAIDHwt cells, wherein MCTs do not impact hyperpolarized pyruvate delivery and lactate production. Collectively, our findings indicated that lower MCT expression was a key contributor to lower hyperpolarized lactate production in NHAIDHmut cells. The SLC16A3 (MCT4) promoter but not SLC16A1 (MCT1) promoter was hypermethylated in NHAIDHmut cells, pointing to possibly different mechanisms mediating reduced MCT expression. Finally analysis of low-grade glioma patient biopsy data from The Cancer Genome Atlas revealed that MCT1 and MCT4 expression was significantly reduced in mutant IDH1 tumors compared to wild-type. Taken together, our study shows that reduced MCT expression is part of the metabolic reprogramming of mutant IDH1 gliomas. This finding could impact treatment and has important implications for metabolic imaging of mutant IDH1 gliomas.

Collaboration


Dive into the Jose L. Izquierdo-Garcia's collaboration.

Top Co-Authors

Avatar

Jesús Ruiz-Cabello

Centro Nacional de Investigaciones Cardiovasculares

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pia Eriksson

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Larry Cai

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge