Network


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

Hotspot


Dive into the research topics where Hayandra F. Nanini is active.

Publication


Featured researches published by Hayandra F. Nanini.


Inflammatory Bowel Diseases | 2014

Overexpression of ATP-activated P2X7 Receptors in the Intestinal Mucosa Is Implicated in the Pathogenesis of Crohnʼs Disease

Adriane R. Neves; Morgana T. Castelo-Branco; Vanessa R. Figliuolo; Claudio Bernardazzi; Fernanda Buongusto; Agnes N. Yoshimoto; Hayandra F. Nanini; Claudia Mara Lara Melo Coutinho; Antonio José V. Carneiro; Robson Coutinho-Silva; Heitor Siffert Pereira de Souza

Background:Extracellular nucleotides released in conditions of cell stress alert the immune system from tissue injury or inflammation. We hypothesized that the P2X7 receptor (P2X7-R) could regulate key elements in inflammatory bowel disease pathogenesis. Methods:Colonoscopy samples obtained from patients with Crohns disease (CD), ulcerative colitis, and controls were used to analyze P2X7-R expression by RT and real-time PCR, immunohistochemistry, and confocal microscopy. Inflammatory response was determined by the levels of cytokines by enzyme-linked immunosorbent assay in cultures of intestinal explants. Apoptosis was determined by the TUNEL assay. P2X7-R−/− C57BL/6 mice were treated with trinitrobenzene sulfonic acid or dextran sulfate sodium (DSS) for inducing colitis. Results:P2X7-R was expressed in higher levels in inflamed CD epithelium and lamina propria, where it colocalizes more with dendritic cells and macrophages. Basal levels of P2X7-R mRNA were higher in CD inflamed mucosa compared with noninflamed CD and controls and were upregulated after interferon-&ggr; in controls. Apoptotic rates were higher in CD epithelium and lamina propria compared with ulcerative colitis and controls. Levels of tumor necrosis factor-&agr;, interleukin (IL)-1&bgr;, and IL-17 were higher, whereas IL-10 was lower in CD compared with controls. Levels of tumor necrosis factor-&agr;-&agr; and interleukin-1&bgr; increased after adenosine-triphosphate and decreased after KN62 treatment in CD. P2X7-R−/− animals did not develop trinitrobenzene sulfonic acid or DSS colitis. Conclusions:The upregulation of P2X7-R in CD inflamed mucosa is consistent with the involvement of purinoceptors in inflammation and apoptosis. These observations may implicate purinergic signaling in the pathogenesis of intestinal inflammation, and the P2X7-R may represent a novel therapeutic target in CD.


Biochimica et Biophysica Acta | 2012

Extracellular ATP induces cell death in human intestinal epithelial cells

Carolina de Oliveira Souza; Giani F. Santoro; Vanessa R. Figliuolo; Hayandra F. Nanini; Heitor Siffert Pereira de Souza; Morgana T. Castelo-Branco; Alessandra Abalo; Mauricio M. Paiva; Claudia Mara Lara Melo Coutinho; Robson Coutinho-Silva

BACKGROUND Extracellular ATP is an endogenous signaling molecule released by various cell types and under different stimuli. High concentrations of ATP released into the extracellular medium activate the P2X7 receptor in most inflammatory conditions. Here, we seek to characterize the effects of ATP in human intestinal epithelial cells and to evaluate morphological changes in these cells in the presence of ATP. METHODS We treated human intestinal epithelial cells with ATP and evaluated the effects of this nucleotide by scanning and transmission electron microscopy analysis and calcium measurements. We used flow cytometry to evaluate apoptosis. We collected human intestinal explants for immunohistochemistry, apoptosis by the TUNEL approach and caspase-3 activity using flow cytometry analyses. We also evaluated the ROS production by flow cytometry and NO secretion by the Griess technique. RESULTS ATP treatment induced changes characteristic of cell death by apoptosis and autophagy but not necrosis in the HCT8 cell line. ATP induced apoptosis in human intestinal explants that showed TUNEL-positive cells in the epithelium and in the lamina propria. The explants exhibited a significant increase of caspase-3 activity when the colonic epithelial cells were incubated with IFN-gamma followed by ATP as compared to control cells. In addition, it was found that antioxidants were able to inhibit both the ROS production and the apoptosis induced by ATP in epithelial cells. GENERAL SIGNIFICANCE The activation of P2X7 receptors by ATP induces apoptosis and autophagy in human epithelial cells, possibly via ROS production, and this effect might have implications for gut inflammatory conditions.


Mediators of Inflammation | 2014

Macrophage P2X7 Receptor Function Is Reduced during Schistosomiasis: Putative Role of TGF-β1

Suellen D’arc Santos Oliveira; Hayandra F. Nanini; Luiz Eduardo Baggio Savio; Mariana Caldas Waghabi; Claudia Silva; Robson Coutinho-Silva

Schistosomiasis is a chronic inflammatory disease whose macrophages are involved in immunopathology modulation. Although P2X7 receptor signaling plays an important role in inflammatory responses mediated by macrophages, no reports have examined the role of P2X7 receptors in macrophage function during schistosomiasis. Thus, we evaluated P2X7 receptor function in peritoneal macrophages during schistosomiasis using an ATP-induced permeabilization assay and measurements of the intracellular Ca2+ concentration. ATP treatment induced significantly less permeabilization in macrophages from S. mansoni-infected mice than in control cells from uninfected animals. Furthermore, P2X7-mediated increases in intracellular Ca2+ levels were also reduced in macrophages from infected mice. TGF-β1 levels were increased in the peritoneal cavity of infected animals, and pretreatment of control macrophages with TGF-β1 reduced ATP-induced permeabilization, mimicking the effect of S. mansoni infection. Western blot and qRT-PCR data showed no difference in P2X7 protein and mRNA between uninfected, infected, and TGF-β1-treated groups. However, immunofluorescence analysis revealed reduced cell surface localization of P2X7 receptors in macrophages from infected and TGF-β1-treated mice compared to controls. Therefore, our data suggest that schistosomiasis reduces peritoneal macrophage P2X7 receptor signaling. This effect is likely due to the fact that infected mice have increased levels of TGF-β1, which reduces P2X7 receptor cell surface expression.


Biochimica et Biophysica Acta | 2017

P2X7 receptor promotes intestinal inflammation in chemically induced colitis and triggers death of mucosal regulatory T cells

Vanessa R. Figliuolo; Luiz Eduardo Baggio Savio; Hanaa Safya; Hayandra F. Nanini; Claudio Bernardazzi; Alessandra Abalo; Heitor Siffert Pereira de Souza; Jean M. Kanellopoulos; Pierre Bobé; Claudia Mara Lara Melo Coutinho; Robson Coutinho-Silva

P2X7 receptor activation contributes to inflammation development in different pathologies. We previously reported that the P2X7 receptor is over-expressed in the gut mucosa of patients with inflammatory bowel disease, and that P2X7 inhibition protects against chemically induced colitis. Here, we investigated in detail the role of the P2X7 receptor in inflammatory bowel disease development, by treating P2X7 knockout (KO) and WT mice with two different (and established) colitis inductors. P2X7 KO mice were protected against gut inflammation induced by 2,4,6-trinitrobenzenesulfonic acid or oxazolone, with no weight loss or gut histological alterations after treatment. P2X7 receptor knockout induced regulatory T cell accumulation in the colon, as evaluated by qRT-PCR for FoxP3 expression and immunostaining for CD90/CD45RBlow. Flow cytometry analysis of mesenteric lymph node cells showed that P2X7 activation (by ATP) triggered regulatory T cell death. In addition, such cells from P2X7 KO mice expressed more CD103, suggesting increased migration of regulatory T cells to the colon (relative to the WT). Our results show that the P2X7 has a key role during inflammation development in inflammatory bowel disease, by triggering the death and retention in the mesenteric lymph nodes of regulatory T cells that would otherwise promote immune system tolerance in the gut.


Life Sciences | 2017

Sulfate-reducing bacteria stimulate gut immune responses and contribute to inflammation in experimental colitis

Vanessa R. Figliuolo; Liliane Martins dos Santos; Alessandra Abalo; Hayandra F. Nanini; Angela Santos; Nilda M. Brittes; Claudio Bernardazzi; Heitor Siffert Pereira de Souza; Leda Quercia Vieira; Robson Coutinho-Silva; Claudia Mara Lara Melo Coutinho

ABSTRACT The intestinal microbiota is critical for mammalian immune system development and homeostasis. Sulfate‐reducing bacteria (SRB) are part of the normal gut microbiota, but their increased levels may contribute to colitis development, likely in association with hydrogen sulfide (H2S) production. Here, we investigated the effects of SRB in the gut immune response in germ‐free mice, and in experimental colitis. After 7 days of colonization with Desulfovibrio indonesiensis or with a human SRB consortium (from patients with colitis), germ‐free mice exhibited alterations in the colonic architecture, with increased cell infiltration in the lamina propria. SRB colonization upregulated the Th17 and Treg profiles of cytokine production/cell activation, in T cells from mesenteric lymph nodes. These alterations were more pronounced in mice colonized with the human SRB consortium, although D. indonesiensis colonization produced higher levels of H2S. Importantly, the colon of C57BL/6 mice with colitis induced by TNBS or oxazolone had increased SRB colonization, and the administration of D. indonesiensis to mice with TNBS‐induced colitis clearly exacerbated the alterations in colonic architecture observed in the established disease, and also increased mouse weight loss. We conclude that SRB contribute to immune response activation in the gut and play an important role in colitis development.


Clinical and Experimental Medicine | 2017

Disruption of the Hedgehog signaling pathway in inflammatory bowel disease fosters chronic intestinal inflammation

Fernanda Buongusto; Claudio Bernardazzi; Agnes N. Yoshimoto; Hayandra F. Nanini; Raquel L. Coutinho; Antonio José V. Carneiro; Morgana T. Castelo-Branco; Heitor S. de Souza

Hedgehog (Hh) signaling is essential for intestinal homeostasis and has been associated with inflammation and tissue repair. We hypothesized that Hh signaling could affect the inflammatory process in inflammatory bowel disease (IBD). For this purpose, colon specimens from the inflamed and non-inflamed mucosa of 15 patients with Crohn’s disease (CD), 15 with ulcerative colitis, and 15 controls were analyzed by immunohistochemistry and real-time PCR. The production and modulation of cytokines were measured by ELISA from culture explants. Apoptosis was assessed by TUNEL and caspase-3 activity assays. Chemotaxis was evaluated using a transwell system. Primary human intestinal and skin fibroblasts were used for analyzing migration and BrdU incorporation. Hh proteins were generally expressed at the superficial epithelium, and a marked reduction was observed in CD. In the lamina propria, Gli-1 predominantly co-localized with vimentin- and alpha-smooth muscle actin-positive cells, with lower levels observed in CD. In colon explants, Hh stimulation resulted in reduction, while blockade increased, TNF α, IL-17, and TGF β levels. Apoptotic rates were higher in inflamed samples, and they increased after Hh blockade. Levels of Gli-1 mRNA were negatively correlated with caspase-3 activity. Hh blockade increased chemoattraction of monocytes. Primary fibroblasts incorporated more BrdU, but migrated less after Hh blockade. These results suggest that Hh signaling provides a negative feedback to the lamina propria, down-regulating inflammatory cytokines, and inhibiting leukocyte migration and fibroblast proliferation, while favoring fibroblast migration. Therefore, Hh signaling is strongly implicated in the pathogenesis of intestinal inflammation, and it may represent a novel therapeutic target for IBD.


Molecular and Cellular Endocrinology | 2016

P2X7 receptor knockout prevents streptozotocin-induced type 1 diabetes in mice

Flávia Sarmento Vieira; Hayandra F. Nanini; Christina Maeda Takiya; Robson Coutinho-Silva


Gastroenterology | 2018

Tu1784 - The Severity of Murine Toxoplasma Gondii -Induced Crohn'slike Ileitis is Modulated by P2X7 Signaling

Hayandra F. Nanini; Aline Cristina Abreu Moreira-Souza; Kivia Q. de Andrade; Luiz Eduardo S. Baggio; Beatriz P. Damasceno; Cynthia Machado Cascabulho; Rossiane C. Vommaro; Robson Coutinho-Silva; Heitor S. de Souza


Gastroenterology | 2018

Su1179 - Protective Effect of Mesenchymal Stem Cells in an Experimental Model of High Risk Intestinal Anastomosis in Rats

Valter Alvarenga; Hayandra F. Nanini; Natalia Bonfa; Beatriz P. Damasceno; Morgana T. Castelo-Branco; Wagner Baetas; Heitor S. de Souza; Alberto Schanaider


Gastroenterology | 2016

Tu1894 Schistosoma Mansoni Coinfection attenuates Murine Toxoplasma Gondii-Induced Crohn's-Like Ileitis by Modulating the Immune Response and the Number of Paneth Cells

Cesonia A. Martinusso; Claudio Bernardazzi; Aline F. Cunha; Jacilene Mesquita; Hayandra F. Nanini; Marta G. Cavalcanti; Heitor S. de Souza

Collaboration


Dive into the Hayandra F. Nanini's collaboration.

Top Co-Authors

Avatar

Robson Coutinho-Silva

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Claudio Bernardazzi

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Morgana T. Castelo-Branco

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heitor Siffert Pereira de Souza

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Vanessa R. Figliuolo

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Agnes N. Yoshimoto

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Antonio José V. Carneiro

Federal University of Rio de Janeiro

View shared research outputs
Researchain Logo
Decentralizing Knowledge