Network


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

Hotspot


Dive into the research topics where Ezequiel J. Tolosa is active.

Publication


Featured researches published by Ezequiel J. Tolosa.


Journal of Biological Chemistry | 2014

The Transcription Factor GLI1 Interacts with SMAD Proteins to Modulate Transforming Growth Factor β-Induced Gene Expression in a p300/CREB-binding Protein-associated Factor (PCAF)-dependent Manner

Monica D. Nye; Luciana L. Almada; Maite G. Fernandez-Barrena; David L. Marks; Sherine F. Elsawa; Anne M. Vrabel; Ezequiel J. Tolosa; Volker Ellenrieder; Martin E. Fernandez-Zapico

Background: The molecular mechanisms mediating the oncogenic activity of the transcription factor GLI1 remain elusive. Results: GLI1 interacts with SMAD factors and PCAF to regulate TGFβ-induced gene expression. Conclusion: These results define a novel epigenetic mechanism underlying the role of GLI1 as an oncogene. Significance: This study increases our understanding of gene expression regulation in cancer cells and its potential impact in tumor development. The biological role of the transcription factor GLI1 in the regulation of tumor growth is well established; however, the molecular events modulating this phenomenon remain elusive. Here, we demonstrate a novel mechanism underlying the role of GLI1 as an effector of TGFβ signaling in the regulation of gene expression in cancer cells. TGFβ stimulates GLI1 activity in cancer cells and requires its transcriptional activity to induce BCL2 expression. Analysis of the mechanism regulating this interplay identified a new transcriptional complex including GLI1 and the TGFβ-regulated transcription factor, SMAD4. We demonstrate that SMAD4 physically interacts with GLI1 for concerted regulation of gene expression and cellular survival. Activation of the TGFβ pathway induces GLI1-SMAD4 complex binding to the BCL2 promoter whereas disruption of the complex through SMAD4 RNAi depletion impairs GLI1-mediated transcription of BCL2 and cellular survival. Further characterization demonstrated that SMAD2 and the histone acetyltransferase, PCAF, participate in this regulatory mechanism. Both proteins bind to the BCL2 promoter and are required for TGFβ- and GLI1-stimulated gene expression. Moreover, SMAD2/4 RNAi experiments showed that these factors are required for the recruitment of GLI1 to the BCL2 promoter. Finally, we determined whether this novel GLI1 transcriptional pathway could regulate other TGFβ targets. We found that two additional TGFβ-stimulated genes, INTERLEUKIN-7 and CYCLIN D1, are dependent upon the intact GLI1-SMAD-PCAF complex for transcriptional activation. Collectively, these results define a novel epigenetic mechanism that uses the transcription factor GLI1 and its associated complex as a central effector to regulate gene expression in cancer cells.


European Journal of Cell Biology | 2016

Sonic hedgehog is a chemotactic neural crest cell guide that is perturbed by ethanol exposure.

Ezequiel J. Tolosa; Martin E. Fernandez-Zapico; Natalia L. Battiato; Roberto A. Rovasio

Our aim was to understand the involvement of Sonic hedgehog (Shh) morphogen in the oriented distribution of neural crest cells (NCCs) toward the optic vesicle and to look for potential disorders of this guiding mechanism after ethanol exposure. In vitro directional analysis showed the chemotactic response of NCCs up Shh gradients and to notochord co-cultures (Shh source) or to their conditioned medium, a response inhibited by anti-Shh antibody, receptor inhibitor cyclopamine and anti-Smo morpholino (MO). Expression of the Ptch-Smo receptor complex on in vitro NCCs was also shown. In whole embryos, the expression of Shh mRNA and protein was seen in the ocular region, and of Ptch, Smo and Gli/Sufu system on cephalic NCCs. Anti-Smo MO or Ptch-mutated plasmid (Ptch1(Δloop2)) impaired cephalic NCC migration/distribution, with fewer cells invading the optic region and with higher cell density at the homolateral mesencephalic level. Beads embedded with cyclopamine (Smo-blocking) or Shh (ectopic signal) supported the role of Shh as an in vivo guide molecule for cephalic NCCs. Ethanol exposure perturbed in vitro and in vivo NCC migration. Early stage embryos treated with ethanol, in a model reproducing Fetal Alcohol Syndrome, showed later disruptions of craniofacial development associated with abnormal in situ expression of Shh morphogen. The results show the Shh/Ptch/Smo-dependent migration of NCCs toward the optic vesicle, with the support of specific inactivation with genetic and pharmacological tools. They also help to understand mechanisms of accurate distribution of embryonic cells and of their perturbation by a commonly consumed teratogen, and demonstrate, in addition to its other known developmental functions, a new biological activity of cellular guidance for Shh.


Cancer Research | 2017

Transcriptional Induction of Periostin by a Sulfatase 2–TGFβ1–SMAD Signaling Axis Mediates Tumor Angiogenesis in Hepatocellular Carcinoma

Gang Chen; Ikuo Nakamura; Renumathy Dhanasekaran; Eriko Iguchi; Ezequiel J. Tolosa; Paola A. Romecin; Renzo E. Vera; Luciana L. Almada; Alexander G. Miamen; Roongruedee Chaiteerakij; Mengtao Zhou; Michael K. Asiedu; Catherine D. Moser; Shaoshan Han; Chunling Hu; Bubu A. Banini; Abdul M. Oseini; Yichun Chen; Yong Fang; Dongye Yang; Hassan M. Shaleh; Shaoqing Wang; Dehai Wu; Tao Song; Ju Seog Lee; Snorri S. Thorgeirsson; Eric Chevet; Vijay H. Shah; Martin E. Fernandez-Zapico; Lewis R. Roberts

Existing antiangiogenic approaches to treat metastatic hepatocellular carcinoma (HCC) are weakly effectual, prompting further study of tumor angiogenesis in this disease setting. Here, we report a novel role for sulfatase 2 (SULF2) in driving HCC angiogenesis. Sulf2-deficient mice (Sulf2 KO) exhibited resistance to diethylnitrosamine-induced HCC and did not develop metastases like wild-type mice (Sulf2 WT). The smaller and less numerous tumors formed in Sulf2 KO mice exhibited a markedly lower microvascular density. In human HCC cells, SULF2 overexpression increased endothelial proliferation, adhesion, chemotaxis, and tube formation in a paracrine fashion. Mechanistic analyses identified the extracellular matrix protein periostin (POSTN), a ligand of αvβ3/5 integrins, as an effector protein in SULF2-induced angiogenesis. POSTN silencing in HCC cells attenuated SULF2-induced angiogenesis and tumor growth in vivo The TGFβ1/SMAD pathway was identified as a critical signaling axis between SULF2 and upregulation of POSTN transcription. In clinical HCC specimens, elevated levels of SULF2 correlated with increased microvascular density, POSTN levels, and relatively poorer patient survival. Together, our findings define an important axis controlling angiogenesis in HCC and a mechanistic foundation for rational drug development. Cancer Res; 77(3); 632-45. ©2016 AACR.


Journal of Biological Chemistry | 2016

Nuclear Factor of Activated T Cells-dependent Down-regulation of the Transcription Factor Glioma-associated Protein 1 (GLI1) Underlies the Growth Inhibitory Properties of Arachidonic Acid.

Andrea Comba; Luciana L. Almada; Ezequiel J. Tolosa; Eriko Iguchi; David L. Marks; Marianela Vara Messler; Renata Silva; Maite G. Fernandez-Barrena; Elisa Enriquez-Hesles; Anne L. Vrabel; Bruno Botta; Lucia Di Marcotulio; V Ellenrieder; Aldo R. Eynard; Maria E. Pasqualini; Martin E. Fernandez-Zapico

Numerous reports have demonstrated a tumor inhibitory effect of polyunsaturated fatty acids (PUFAs). However, the molecular mechanisms modulating this phenomenon are in part poorly understood. Here, we provide evidence of a novel antitumoral mechanism of the PUFA arachidonic acid (AA). In vivo and in vitro experiments showed that AA treatment decreased tumor growth and metastasis and increased apoptosis. Molecular analysis of this effect showed significantly reduced expression of a subset of antiapoptotic proteins, including BCL2, BFL1/A1, and 4-1BB, in AA-treated cells. We demonstrated that down-regulation of the transcription factor glioma-associated protein 1 (GLI1) in AA-treated cells is the underlying mechanism controlling BCL2, BFL1/A1, and 4-1BB expression. Using luciferase reporters, chromatin immunoprecipitation, and expression studies, we found that GLI1 binds to the promoter of these antiapoptotic molecules and regulates their expression and promoter activity. We provide evidence that AA-induced apoptosis and down-regulation of antiapoptotic genes can be inhibited by overexpressing GLI1 in AA-sensitive cells. Conversely, inhibition of GLI1 mimics AA treatments, leading to decreased tumor growth, cell viability, and expression of antiapoptotic molecules. Further characterization showed that AA represses GLI1 expression by stimulating nuclear translocation of NFATc1, which then binds the GLI1 promoter and represses its transcription. AA was shown to increase reactive oxygen species. Treatment with antioxidants impaired the AA-induced apoptosis and down-regulation of GLI1 and NFATc1 activation, indicating that NFATc1 activation and GLI1 repression require the generation of reactive oxygen species. Collectively, these results define a novel mechanism underlying AA antitumoral functions that may serve as a foundation for future PUFA-based therapeutic approaches.


Journal of Immunology | 2015

Modulation of the IL-6 Receptor α Underlies GLI2-Mediated Regulation of Ig Secretion in Waldenström Macroglobulinemia Cells

David A. Jackson; Timothy D. Smith; Nansalmaa Amarsaikhan; Weiguo Han; Matthew S. Neil; Shannon K. Boi; Anne M. Vrabel; Ezequiel J. Tolosa; Luciana L. Almada; Martin E. Fernandez-Zapico; Sherine F. Elsawa

Ig secretion by terminally differentiated B cells is an important component of the immune response to foreign pathogens. Its overproduction is a defining characteristic of several B cell malignancies, including Waldenström macroglobulinemia (WM), where elevated IgM is associated with significant morbidity and poor prognosis. Therefore, the identification and characterization of the mechanisms controlling Ig secretion are of great importance for the development of future therapeutic approaches for this disease. In this study, we define a novel pathway involving the oncogenic transcription factor GLI2 modulating IgM secretion by WM malignant cells. Pharmacological and genetic inhibition of GLI2 in WM malignant cells resulted in a reduction in IgM secretion. Screening for a mechanism identified the IL-6Rα (gp80) subunit as a downstream target of GLI2 mediating the regulation of IgM secretion. Using a combination of expression, luciferase, and chromatin immunoprecipitation assays we demonstrate that GLI2 binds to the IL-6Rα promoter and regulates its activity as well as the expression of this receptor. Additionally, we were able to rescue the reduction in IgM secretion in the GLI2 knockdown group by overexpressing IL-6Rα, thus defining the functional significance of this receptor in GLI2-mediated regulation of IgM secretion. Interestingly, this occurred independent of Hedgehog signaling, a known regulator of GLI2, as manipulation of Hedgehog had no effect on IgM secretion. Given the poor prognosis associated with elevated IgM in WM patients, components of this new signaling axis could be important therapeutic targets.


Journal of Biological Chemistry | 2016

Gli transcription factors mediate the oncogenic transformation of prostate basal cells induced by a Kras-androgen receptor axis

Meng Wu; Ezequiel J. Tolosa; Renzo E. Vera; Qianjin Li; Sungjin Kim; Yongjie Ma; Demetri D. Spyropoulos; Zanna Beharry; Jiaoti Huang; Martin E. Fernandez-Zapico; Houjian Cai

Although the differentiation of oncogenically transformed basal progenitor cells is one of the key steps in prostate tumorigenesis, the mechanisms mediating this cellular process are still largely unknown. Here we demonstrate that an expanded p63+ and CK5+ basal/progenitor cell population, induced by the concomitant activation of oncogenic Kras(G12D) and androgen receptor (AR) signaling, underwent cell differentiation in vivo. The differentiation process led to suppression of p63-expressing cells with a decreased number of CK5+ basal cells but an increase of CK8+ luminal tumorigenic cells and revealed a hierarchal lineage pattern consisting of p63+/CK5+ progenitor, CK5+/CK8+ transitional progenitor, and CK8+ differentiated luminal cells. Further analysis of the phenotype showed that Kras-AR axis-induced tumorigenesis was mediated by Gli transcription factors. Combined blocking of the activators of this family of proteins (Gli1 and Gli2) inhibited the proliferation of p63+ and CK5+ basal/progenitor cells and development of tumors. Finally, we identified that Gli1 and Gli2 exhibited different functions in the regulation of p63 expression or proliferation of p63+ cells in Kras-AR driven tumors. Gli2, but not Gli1, transcriptionally regulated the expression levels of p63 and prostate sphere formation. Our study provides evidence of a novel mechanism mediating pathological dysregulation of basal/progenitor cells through the differential activation of the Gli transcription factors. Also, these findings define Gli proteins as new downstream mediators of the Kras-AR axis in prostate carcinogenesis and open a potential therapeutic avenue of targeting prostate cancer progression by inhibiting Gli signaling.


Molecular Therapy | 2016

413. Effect of Adenoviral Death Protein on NIS-Based Iodine Therapy and Imaging for Pancreatic Cancer

Lisa Koodie; Eriko Kawakami; Ben Eidenschink; Jordan Sell; Chris LaRocca; Kari Jacobsen; Jessica Ryvlin; Ezequiel J. Tolosa; John C. Morris; Martin E. Fernandez-Zapico; Julia Davydova

In this study, we assess our novel Oncolytic Adenovirus (OAd) expressing a dual function therapeutic and imaging transgene, the sodium-iodide symporter (NIS) as a promising alternative for pancreatic cancer treatment. We hypothesized that NIS expression in pancreatic cancer will induce uptake of radioiodine and allow noninvasive SPECT/CT imaging with 123I. We designed our OAd-NIS to overcome the low efficacy of previous vectors. These viruses have a modified Ad5/Ad3 fiber-knob shown to improve the poor transduction of pancreatic cancer cells. Viral replication is controlled under the Cox2 promoter allowing specific delivery of viral genes, and most of adenoviral E3 genes are deleted and replaced with NIS and Adenovirus Death Protein (ADP) genes. We have previously demonstrated significantly improved ability of ADP-overexpressing OAds to enhance viral release and cytolytic activity. Although improved oncolysis enhances viral release and improve the therapeutic effect in solid tumors, it can potentially reduce radioiodide tracer uptake. To test this possibility we designed and compared identical vectors with ADP (OAd-NIS-ADP) and without ADP (OAd-NIS-noADP) in vitro and in vivo. We first compared virus killing ability in pancreatic cancer cell lines. The ADP+ vector was significantly more cytolytic that no-ADP counterpart and showed improved viral replication and spread. Cox2-controlled OAd did not produce cell death in Cox2-negative control, confirming selectivity. We next assessed OAds ability to induce NIS protein expression and radioiodine uptake. NIS-OAds efficiently produce glycosylated NIS multimers as early as 2-days post infection. Infection with no-ADP vectors resulted in a significantly greater radioiodine uptake (125I) compared to ADP+ viruses. Importantly, ex vivo uptake test in human patient tissues confirmed the high level of NIS in pancreatic adenocarcinoma samples, and revealed no 125I uptake in normal pancreas. We further assessed the ability of our vectors to visualize human pancreatic cancer xenografts in a mouse model by monitoring 99mTcO4− accumulation with SPECT-CT. The OAd-NIS-noADP showed an earlier and more sustained radioisotope uptake when compared to ADP+ supporting its use as a more sensitive diagnostic tool for pancreatic cancer. Of contrary, the ADP+ vector significantly outperformed OAd-NIS-noADP in tumor shrinkage. These results suggest that while OAd-NIS-noADP produces a greater radiotracer uptake and can be used as a diagnostic tool, its ADP+ counterpart results in a better therapeutic effect when applied as a monotherapy. It is now essential to estimate the therapeutic ability of both vectors upon combination with I-131 (ongoing study). Further investigation will include the imaging and therapeutic studies in patient-derived xenografts. Ultimately, the goal of our research is to design a multimodal therapy with radiation and oncolytic virus for diagnosis and therapy of cancers.


Gut | 2018

Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming

Marina Koutsioumpa; Maria Hatziapostolou; Christos Polytarchou; Ezequiel J. Tolosa; Luciana L. Almada; Swapna Mahurkar-Joshi; Jennifer L. Williams; Ana B. Tirado-Rodriguez; Sara Huerta-Yepez; Dimitrios Karavias; Helen P. Kourea; George A. Poultsides; Kevin Struhl; David W. Dawson; Timothy R. Donahue; Martin E. Fernandez-Zapico; Dimitrios Iliopoulos

Objective Despite advances in the identification of epigenetic alterations in pancreatic cancer, their biological roles in the pathobiology of this dismal neoplasm remain elusive. Here, we aimed to characterise the functional significance of histone lysine methyltransferases (KMTs) and demethylases (KDMs) in pancreatic tumourigenesis. Design DNA methylation sequencing and gene expression microarrays were employed to investigate CpG methylation and expression patterns of KMTs and KDMs in pancreatic cancer tissues versus normal tissues. Gene expression was assessed in five cohorts of patients by reverse transcription quantitative-PCR. Molecular analysis and functional assays were conducted in genetically modified cell lines. Cellular metabolic rates were measured using an XF24-3 Analyzer, while quantitative evaluation of lipids was performed by liquid chromatography-mass spectrometry (LC-MS) analysis. Subcutaneous xenograft mouse models were used to evaluate pancreatic tumour growth in vivo. Results We define a new antitumorous function of the histone lysine (K)-specific methyltransferase 2D (KMT2D) in pancreatic cancer. KMT2D is transcriptionally repressed in human pancreatic tumours through DNA methylation. Clinically, lower levels of this methyltransferase associate with poor prognosis and significant weight alterations. RNAi-based genetic inactivation of KMT2D promotes tumour growth and results in loss of H3K4me3 mark. In addition, KMT2D inhibition increases aerobic glycolysis and alters the lipidomic profiles of pancreatic cancer cells. Further analysis of this phenomenon identified the glucose transporter SLC2A3 as a mediator of KMT2D-induced changes in cellular, metabolic and proliferative rates. Conclusion Together our findings define a new tumour suppressor function of KMT2D through the regulation of glucose/fatty acid metabolism in pancreatic cancer.


Pancreatology | 2016

Effect of adenoviral death protein on NIS-based iodine therapy and imaging for pancreatic cancer

Lisa Koodie; Ben Eidenschink; Jordan Sell; Chris LaRocca; Kari Jacobsen; Jessica Ryvlin; Ezequiel J. Tolosa; John C. Morris; Martin Fernandez-Zappico; Julia Davydova; Masato Yamamoto


Oncotarget | 2015

Overexpression of mutant Ptch in rhabdomyosarcomas is associated with promoter hypomethylation and increased Gli1 and H3K4me3 occupancy

Frauke Nitzki; Ezequiel J. Tolosa; Nicole Cuvelier; Anke Frommhold; Gabriela Salinas-Riester; Steven A. Johnsen; Martin E. Fernandez-Zapico; Heidi Hahn

Collaboration


Dive into the Ezequiel J. Tolosa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa Koodie

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Sherine F. Elsawa

Northern Illinois University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge