Network


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

Hotspot


Dive into the research topics where Claudia Gabriela Pellizas is active.

Publication


Featured researches published by Claudia Gabriela Pellizas.


The FASEB Journal | 2008

Control of dendritic cell maturation and function by triiodothyronine

Iván D. Mascanfroni; María del Mar Montesinos; Sebastián Susperreguy; Laura Cervi; Juan M. Ilarregui; Vanesa D. Ramseyer; Ana M. Masini-Repiso; Héctor M. Targovnik; Gabriel A. Rabinovich; Claudia Gabriela Pellizas

Accumulating evidence indicates a functional crosstalk between immune and endocrine mechanisms in the modulation of innate and adaptive immunity. However, the impact of thyroid hormones (THs) in the initiation of adaptive immune responses has not yet been examined. Here we investigated the presence of thyroid hormone receptors (TRs) and the impact of THs in the physiology of mouse dendritic cells (DCs), specialized antigen‐presenting cells with the unique capacity to fully activate naive T cells and orchestrate adaptive immunity. Both immature and lipopolysaccharide‐matured bone marrow‐derived DCs expressed TRs at mRNA and protein levels, showing a preferential cytoplasmic localization. Remarkably, physiological levels of triiodothyronine (T3) stimulated the expression of DC maturation markers (major histocompatability complex II, CD80, CD86, and CD40), markedly in‐creased the secretion of interleukin‐12, and stimulated the ability of DCs to induce naive T cell proliferation and IFN‐γ production in allogeneic T cell cultures. Analysis of the mechanisms involved in these effects revealed the ability of T3 to influence the cytoplasmic‐nuclear shuttling of nuclear factor‐κB on primed DCs. Our study provides the first evidence for the presence of TRs on bone marrow‐derived DCs and the ability of THs to regulate DC maturation and function. These results have profound implications in immunopathology, including cancer and autoimmune manifestations of the thyroid gland at the crossroads of the immune and endocrine systems. Mascanfroni, I., Montesinos, M., Susperreguy, S., Cervi, L., Ilarregui, J. M., Ramseyer, V. D., Masini‐Repiso, A. M., Targovnik, H. M., Rabinovich, G. A., Pellizas, C. G. Control of dendritic cell maturation and function by triiodothyronine. FASEB J. 22, 1032–1042 (2008)


Endocrinology | 2009

Functional Toll-Like Receptor 4 Conferring Lipopolysaccharide Responsiveness Is Expressed in Thyroid Cells

Juan Pablo Nicola; María Laura Vélez; Ariel Maximiliano Lucero; Laura Fozzatti; Claudia Gabriela Pellizas; Ana M. Masini-Repiso

Lipopolysaccharide (LPS), a glycolipid found in the cell wall of Gram-negative bacteria, exerts pleiotropic biological effects in different cell types. LPS is mainly recognized by the Toll-like receptor (TLR) 4/MD2/Cluster of differentiation 14 complex (CD14). We previously demonstrated that LPS produced a direct action on thyroid cells, including up-regulation of thyroglobulin gene expression. This work aimed to study further the effect of LPS on thyroid function and to elucidate the mechanism by which LPS is recognized by the thyroid cell. We could detect the transcript and protein expression of TLR4, MD2, and CD14 in thyroid cells, and that these proteins are localized at the plasma membrane. The sodium iodide symporter (NIS) is the transporter involved in the iodide uptake, the first step in thyroid hormonogenesis. We demonstrated that LPS increases the TSH-induced iodide uptake and NIS protein expression. The LPS agonist lipid A reproduced LPS effect, whereas the LPS antagonist, polymyxin B, abrogated it. By the use of anti-TLR4 blocking antibodies and the transient expression of TLR4 dominant-negative forms, we evidenced the involvement of TLR4 in the LPS action. The enrichment of TLR4 expressing Fisher rat thyroid cell line-5 (FRTL-5) cells confirmed that TLR4 confers LPS responsiveness to thyroid cells. In conclusion, we revealed for the first time that all the components of the LPS receptor complex are expressed in thyroid cells. Evidence that the effects of LPS on rodent thyroid function involve TLR4-induced signaling was obtained. The fact that thyroid cells are able to recognize and respond to LPS supports a role of the endotoxin as a potential modifier of thyroid function.


Journal of Biological Chemistry | 2010

Nuclear Factor (NF)-κB-dependent Thyroid Hormone Receptor β1 Expression Controls Dendritic Cell Function via Akt Signaling

Ivan D. Mascanfroni; María del Mar Montesinos; Vanina A. Alamino; Sebastián Susperreguy; Juan Pablo Nicola; Juan M. Ilarregui; Ana M. Masini-Repiso; Gabriel A. Rabinovich; Claudia Gabriela Pellizas

Despite considerable progress in our understanding of the interplay between immune and endocrine systems, the role of thyroid hormones and their receptors in the control of adaptive immunity is still uncertain. Here, we investigated the role of thyroid hormone receptor (TR) β1 signaling in modulating dendritic cell (DC) physiology and the intracellular mechanisms underlying these immunoregulatory effects. Exposure of DCs to triiodothyronine (T3) resulted in a rapid and sustained increase in Akt phosphorylation independently of phosphatidylinositol 3-kinase activation, which was essential for supporting T3-induced DC maturation and interleukin (IL)-12 production. This effect was dependent on intact TRβ1 signaling as small interfering RNA-mediated silencing of TRβ1 expression prevented T3-induced DC maturation and IL-12 secretion as well as Akt activation and IκB-ϵ degradation. In turn, T3 up-regulated TRβ1 expression through mechanisms involving NF-κB, suggesting an autocrine regulatory loop to control hormone-dependent TRβ1 signaling. These findings were confirmed by chromatin immunoprecipitation analysis, which disclosed a new functional NF-κB consensus site in the promoter region of the TRB1 gene. Thus, a T3-induced NF-κB-dependent mechanism controls TRβ1 expression, which in turn signals DCs to promote maturation and function via an Akt-dependent but PI3K-independent pathway. These results underscore a novel unrecognized target that regulates DC maturation and function with critical implications in immunopathology at the cross-roads of the immune-endocrine circuits.


Molecular Endocrinology | 2010

NF-κB p65 Subunit Mediates Lipopolysaccharide-Induced Na+/I- Symporter Gene Expression by Involving Functional Interaction with the Paired Domain Transcription Factor Pax8

Juan Pablo Nicola; Magalí Nazar; Iván D. Mascanfroni; Claudia Gabriela Pellizas; Ana M. Masini-Repiso

The Gram-negative bacterial endotoxin lipopolysaccharide (LPS) elicits a variety of biological responses. Na(+)/I(-) symporter (NIS)-mediated iodide uptake is the main rate-limiting step in thyroid hormonogenesis. We have recently reported that LPS stimulates TSH-induced iodide uptake. Here, we further analyzed the molecular mechanism involved in the LPS-induced NIS expression in Fisher rat thyroid cell line 5 (FRTL-5) thyroid cells. We observed an increase in TSH-induced NIS mRNA expression in a dose-dependent manner upon LPS treatment. LPS enhanced the TSH-stimulated NIS promoter activity denoting the NIS-upstream enhancer region (NUE) as responsible for the stimulatory effects. We characterized a novel putative conserved kappaB site for the transcription factor nuclear factor-kappaB (NF-kappaB) within the NUE region. NUE contains two binding sites for the transcription factor paired box 8 (Pax8), main regulator of NIS transcription. A physical interaction was observed between the NF-kappaB p65 subunit and paired box 8 (Pax8), which appears to be responsible for the synergic effect displayed by these transcription factors on NIS gene transcription. Moreover, functional blockage of NF-kappaB signaling and site-directed mutagenesis of the kappaB cis-acting element abrogated LPS stimulation. Silencing expression of p65 confirmed its participation as an effector of LPS-induced NIS stimulation. Furthermore, chromatin immunoprecipitation corroborated that NIS is a novel target gene for p65 transactivation in response to LPS. Moreover, we were able to corroborate the LPS-stimulatory effect on thyroid cells in vivo in LPS-treated rats, supporting that thyrocytes are capable of responding to systemic infections. In conclusion, our results reveal a new mechanism involving p65 in the LPS-induced NIS expression, denoting a novel aspect in thyroid cell differentiation.


Journal of Cellular Physiology | 2011

Cooperative nongenomic and genomic actions on thyroid hormone mediated-modulation of T cell proliferation involve up-regulation of thyroid hormone receptor and inducible nitric oxide synthase expression.

María Laura Barreiro Arcos; Helena A. Sterle; Maria A. Paulazo; Eduardo Valli; Alicia Juana Klecha; Blanca Isse; Claudia Gabriela Pellizas; Ricardo N. Farías; Graciela Cremaschi

Thyroid hormones (THs) exert a broad range of actions on development, growth, and cell differentiation by both genomic and nongenomic mechanisms. THs regulate lymphocyte function, but the participation of nongenomic actions is still unknown. Here the contribution of both genomic and nongenomic effects on TH‐induced division of T cells was studied by using free and noncell permeable THs coupled to agarose (TH‐ag). THs‐ag led to cell division, but to a lesser extent than free hormones. THs induced nongenomically the rapid translocation of protein kinase C (PKC) ζ isoform to cell membranes, extracellular‐signal‐regulated kinases (ERK1/2) phosphorylation and nuclear factor‐κB (NF‐κB) activation. The signaling cascade include sphingomielinases acting up‐stream the activation of PKCζ isoform, while ERK and NF‐κB are activated downstream this PKC isoenzyme. Both free and THs‐ag increased the protein and mRNA levels of TH nuclear receptor TRα1, while only free hormones incremented the inducible NOS gene and protein levels as well as a calcium independent NOS activity. Both effects were blunted by PKCζ inhibition. These results indicate that THs, by triggering a nongenomic signaling cascade that involves Smases‐mediated activation of PKCζ, lead to ERK 1/2 and NF‐κB activation and to the genomic increase of TRs and the inducible nitric oxide synthase protein and mRNA levels, improving T lymphocyte proliferation. These finding not only contribute to the understanding of the mechanisms involved in TH modulation of lymphocyte physiology, but would also point out for the first time the interplay between genomic and nongenomic TH actions in T cells. J. Cell. Physiol. 226: 3208–3218, 2011.


Histochemical Journal | 2001

Behaviour of a somatotroph population under a growth hormone releasing peptide treatment

Mónica Bonaterra; Ana Lucía De Paul; Claudia Gabriela Pellizas; Agustín Aoki; Alicia Inés Torres

In this investigation, we studied the effects of Momany peptide (GHRP-5), on somatotroph secretory activity. Acute and chronic administration of GHRP-5 provokes a significant release of growth hormone that can be closely correlated with ultrastructural changes in somatotroph populations. After 3, 5 and 7 days of GHRP-5 treatment, two somatotroph cell subpopulations coexist. One of them has an enhanced secretory activity and the other presents a quiescent appearance. Therefore, pituitary growth hormone content was not affected in the first seven days of GHRP-5 treatment. After 14 days, there was a significant depletion of growth hormone pituitary content coincident with the highest levels of serum growth hormone. These results concur with the surge of a new hyperactive somatotroph subtype characterised by numerous immature secretory granules that are discharged bypassing the maturation step. Acute and chronic treatments caused no changes in somatotroph cell density, the area immunostained for growth hormone and the levels of total mRNA for transcription factor pit-1.The results of pituitary cell cultures incubated with specific blockers for different signalling pathways demonstrated an involvement of the phospholipase C–inositol phosphate system in GHRP-5 stimulated somatotroph secretion.GHRP-5 treatment enhanced significantly the release of growth hormone, thereby eliciting ultrastructural modifications in somatotrophs that can be correlated with an increased secretory activity devoid of cell density changes.


Histochemistry and Cell Biology | 2008

Extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinases (MMPs) as regulators of tumor–host interaction in a spontaneous metastasis model in rats

Ana Carolina Donadio; María Mónica Remedi; Sebastián Susperreguy; Silvia Frede; Mónica Gilardoni; Yi Tang; Claudia Gabriela Pellizas; Li Yan

EMMPRIN has a role in invasion and metastasis through the induction of MMPs and the consequent modulation of cell-substrate and cell–cell adhesion processes. The present study evaluates the expression of EMMPRIN protein and MMP-2/9 activity in tumor and parenchymal cells in a spontaneous metastasis model in rats. Moreover, we explore the regulation of EMMPRIN and MMP-9 by tumor-epithelial cell interactions in vitro. By zymography, we observed an increased proMMP-9 expression in both metastasized liver and spleen samples from tumor bearing rats. Immunohistochemical studies showed EMMPRIN-positive tumor cells in tumor biopsies as well as in spleen and liver samples from tumor bearing rats. Interestingly, a significant increase in EMMPRIN expression in hepatic cells was also detected. The regulation of EMMPRIN expression in tumor and liver cells in response to tumor–host interaction was investigated in vitro through a tumor cell line culture on extracellular matrix (ECM) molecules or in co-culture with normal rat liver cells (BRL3A cells). No significant changes in EMMPRIN expression were detected in tumor cells cultured on ECM molecules. On the other hand, EMMPRIN protein and MMP-9 mRNA expression were induced in BRL3A cells. The increase in EMMPRIN expression in BRL3A cells was inhibited by an anti-EMMPRIN antibody. These results reinforce the main role of EMMPRIN mediating tumor–host interactions that may evolve new opportunities for therapeutic interventions.


Endocrinology | 2012

Thyroid Peroxidase Gene Expression Is Induced by Lipopolysaccharide Involving Nuclear Factor (NF)-κB p65 Subunit Phosphorylation

Magalí Nazar; Juan Pablo Nicola; María Laura Vélez; Claudia Gabriela Pellizas; Ana M. Masini-Repiso

Thyroid peroxidase (TPO), a tissue-specific enzyme expressed in differentiated thyroid follicular cells, is a major antigen that has been linked to autoimmune thyroid disease. We have previously reported the functional expression of the lipopolysaccharide (LPS) receptor Toll-like receptor 4 on thyroid follicular cells. Here we investigated the effect of LPS in TPO expression and analyzed the mechanisms involved. We found a dose-dependent enhancement of TSH-induced TPO expression in response to LPS stimulation. EMSAs demonstrated that LPS treatment increased thyroid transcription factor-1 and -2 binding to the B and Z regions of TPO promoter, respectively. Moreover, LPS increased TSH-stimulated TPO promoter activity. Using bioinformatic analysis, we identified a conserved binding site for transcription nuclear factor-κB (NF-κB) in the TPO promoter. Chemical inhibition of NF-κB signaling and site-directed mutagenesis of the identified κB-cis-acting element abolished LPS stimulation. Furthermore, chromatin immunoprecipitation assays confirmed that TPO constitutes a novel NF-κB p65 subunit target gene in response to LPS. Additionally, our results indicate that p65 phosphorylation of serine 536 constitutes an essential step in the p65-dependent, LPS-induced transcriptional expression of TPO. In conclusion, here we demonstrated that LPS increases TPO expression, suggesting a novel mechanism involved in the regulation of a major thyroid autoantigen. Our results provide new insights into the potential effects of infectious processes on thyroid homeostasis.


Steroids | 2012

Dexamethasone counteracts the immunostimulatory effects of triiodothyronine (T3) on dendritic cells

María del Mar Montesinos; Vanina A. Alamino; Iván D. Mascanfroni; Sebastián Susperreguy; Nicolás Gigena; Ana M. Masini-Repiso; Gabriel A. Rabinovich; Claudia Gabriela Pellizas

Glucocorticoids (GCs) are widely used as anti-inflammatory and immunosuppressive agents. Several studies have indicated the important role of dendritic cells (DCs), highly specialized antigen-presenting and immunomodulatory cells, in GC-mediated suppression of adaptive immune responses. Recently, we demonstrated that triiodothyronine (T3) has potent immunostimulatory effects on bone marrow-derived mouse DCs through a mechanism involving T3 binding to cytosolic thyroid hormone receptor (TR) β1, rapid and sustained Akt activation and IL-12 production. Here we explored the impact of GCs on T3-mediated DC maturation and function and the intracellular events underlying these effects. Dexamethasone (Dex), a synthetic GC, potently inhibited T3-induced stimulation of DCs by preventing the augmented expression of maturation markers and the enhanced IL-12 secretion through mechanisms involving the GC receptor. These effects were accompanied by increased IL-10 levels following exposure of T3-conditioned DCs to Dex. Accordingly, Dex inhibited the immunostimulatory capacity of T3-matured DCs on naive T-cell proliferation and IFN-γ production while increased IL-10 synthesis by allogeneic T cell cultures. A mechanistic analysis revealed the ability of Dex to dampen T3 responses through modulation of Akt phosphorylation and cytoplasmic-nuclear shuttling of nuclear factor-κB (NF-κB). In addition, Dex decreased TRβ1 expression in both immature and T3-maturated DCs through mechanisms involving the GC receptor. Thus GCs, which are increased during the resolution of inflammatory responses, counteract the immunostimulatory effects of T3 on DCs and their ability to polarize adaptive immune responses toward a T helper (Th)-1-type through mechanisms involving, at least in part, NF-κB- and TRβ1-dependent pathways. Our data provide an alternative mechanism for the anti-inflammatory effects of GCs with critical implications in immunopathology at the cross-roads of the immune-endocrine circuits.


Histochemistry and Cell Biology | 2008

Thyroid hormone receptor α1–β1 expression in epididymal epithelium from euthyroid and hypothyroid rats

Ana Lucía De Paul; Jorge Humberto Mukdsi; Claudia Gabriela Pellizas; María del Mar Montesinos; Silvina Gutiérrez; Sebastián Susperreguy; Alberto Del Río; Cristina A. Maldonado; Alicia Inés Torres

The objectives of the present work were to assess whether epithelial cells from the different segments of epididymis express TRα1–β1 isoforms, to depict its subcellular immunolocalization and to evaluate changes in their expression in rats experimentally submitted to a hypothyroid state by injection of 131I. In euthyroid and hypothyroid groups, TR protein was expressed in epididymal epithelial cells, mainly in the cytoplasmic compartment while only a few one showed a staining in the nucleus as well. A similar TR immunostaining pattern was detected in the different segments of the epididymis. In hypothyroid rats, the number of TR-immunoreactive epithelial cells as well as the intensity of the cytoplasmic staining significantly increased in all sections analyzed. In consonance to the immunocytochemical analysis, the expression of TRα1–β1 isoforms, assessed by Western blot revealed significantly higher levels of TR in cytosol compared to the nuclear fractions. Furthermore, TR expression of both α1 and β1 isoforms and their mRNA levels were increased by the hypothyroid state. The immuno-electron-microscopy showed specific reaction for TR in principal cells associated with eucromatin, cytosolic matrix and mitochondria. The differences in expression levels assessed in control and thyroidectomized rats ascertain a specific function of TH on this organ.

Collaboration


Dive into the Claudia Gabriela Pellizas's collaboration.

Top Co-Authors

Avatar

Ana M. Masini-Repiso

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aldo H. Coleoni

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar

Iván D. Mascanfroni

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar

Juan Pablo Nicola

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar

Sebastián Susperreguy

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magalí Nazar

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar

María Laura Vélez

National University of Cordoba

View shared research outputs
Top Co-Authors

Avatar

Alicia Inés Torres

National University of Cordoba

View shared research outputs
Researchain Logo
Decentralizing Knowledge