Network


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

Hotspot


Dive into the research topics where Rudimar Luiz Frozza is active.

Publication


Featured researches published by Rudimar Luiz Frozza.


Neurobiology of Disease | 2006

Protective effect of resveratrol against oxygen-glucose deprivation in organotypic hippocampal slice cultures : Involvement of PI3-K pathway

Lauren Lúcia Zamin; Patrícia Dillenburg-Pilla; Ricardo Argenta-Comiran; Ana Paula Horn; Fabrício Simão; Melissa Calegaro Nassif; Daniéli Gerhardt; Rudimar Luiz Frozza; Christianne Gazzana Salbego

Here we investigated the neuroprotective effect of resveratrol in an in vitro model of ischemia. We used organotypic hippocampal cultures exposed to oxygen-glucose deprivation (OGD). In OGD-vehicle exposed cultures, about 46% of the hippocampus was labeled with PI, indicating a robust percentage of cell death. When cultures were treated with resveratrol 10, 25 and 50 microM, the cell death was reduced to 22, 20 and 13% respectively. To elucidate a possible mechanism by which resveratrol exerts its neuroprotective effect, we investigated the phosphoinositide3-kinase (PI3-k) pathway using LY294002 (5 microM) and mitogen-activated protein kinase (MAPK) using PD98059 (20 microM). The resveratrol (50 microM) neuroprotection was prevented by LY294002 but was not by PD98059. Immunoblotting revealed that resveratrol 50 microM induced the phosphorylation/activation of Akt and extracellular signal-regulated kinase-1 and -2 (ERK1/2) and the phosphorylation/inactivation of glycogen synthase kinase-3beta (GSK-3beta). Our results suggest that PI3-k/Akt pathway are involved in the neuroprotective effect of resveratrol.


Journal of Pineal Research | 2010

Amyloid‐β neurotoxicity in organotypic culture is attenuated by melatonin: involvement of GSK‐3β, tau and neuroinflammation

Juliana Bender Hoppe; Rudimar Luiz Frozza; Ana Paula Horn; Ricardo Argenta Comiran; Andressa Bernardi; Maria M. Campos; Ana Maria Oliveira Battastini; Christianne Gazzana Salbego

Abstract:  Alzheimer’s disease (AD) is a progressive neurodegenerative disorder marked by accumulation of extracellular deposits of amyloid‐β (Aβ) peptide in brain regions that are important for memory and cognition. The buildup of Aβ aggregates in the AD is followed by the formation of intracellular neurofibrillary tangles and activation of neuroinflammatory reactions. The present study investigated whether melatonin possesses a neuroprotective effect against Aβ‐induced toxicity. For this purpose, organotypic hippocampal slices were cultured and exposed to 25 μm of Aβ25–35 in the absence or in the presence of melatonin (25, 50, or 100 μm). In addition, the authors have investigated the involvement of GSK‐3β, tau protein, astroglial, and microglial activation, and cytokine levels in the melatonin protection against Aβ‐induced neurotoxicity. Melatonin prevented the cell damage in hippocampus induced by the exposure to Aβ25–35. In addition, melatonin significantly reduced the activation of GSK‐3β, the phosphorylation of tau protein, the glial activation and the Aβ‐induced increase of TNF‐α and IL‐6 levels. On the basis of these findings, we speculate that melatonin may provide an effective therapeutic strategy for AD, by attenuating Aβ‐induced phosphorylation of tau protein, and preventing GSK‐3β activation and neuroinflammation.


Neurochemical Research | 2005

Estradiol Protects Against Oxygen and Glucose Deprivation in Rat Hippocampal Organotypic Cultures and Activates Akt and Inactivates GSK-3β

Helena Iturvides Cimarosti; Lauren Lúcia Zamin; Rudimar Luiz Frozza; Melissa Calegaro Nassif; Ana Paula Horn; Alexandre Altino Tavares; Carlos Alexandre Netto; Christianne Gazzana Salbego

Here we investigated the neuroprotective effect of 17β-estradiol in an in vitro model of ischemia. We used organotypic hippocampal slice cultures, acute or chronically treated with 17β-estradiol (10 nM), and exposed to oxygen and glucose deprivation (OGD). Cellular death was quantified by measuring uptake of propidium iodide (PI), a marker of dead cells. In OGD exposed cultures, treated only with vehicle, about 70% of the CA1 area of hippocampus was labeled with PI, indicating a great percentage of cellular death. When cultures were treated with 17β-estradiol (acute or chronically), this cellular death was reduced to 15%. This effect was prevented by LY294002 but was not by PD98059. Immunoblotting revealed that both, chronic and acute, treatments with 17β-estradiol induced the phosphorylation/activation of Akt and the phosphorylation/inactivation of GSK-3β. Our results show a clear neuroprotective effect of 17β-estradiol and suggest that this effect could involve PI3-K pathway.


International Journal of Nanomedicine | 2012

Indomethacin-loaded lipid-core nanocapsules reduce the damage triggered by Aβ1-42 in Alzheimer’s disease models

Andressa Bernardi; Rudimar Luiz Frozza; André Meneghetti; Juliana Bender Hoppe; Ana Maria Oliveira Battastini; Adriana Raffin Pohlmann; Silvia Stanisçuaski Guterres; Christianne Gazzana Salbego

Neuroinflammation, characterized by the accumulation of activated microglia and reactive astrocytes, is believed to modulate the development and/or progression of Alzheimer’s disease (AD). Epidemiological studies suggesting that nonsteroidal anti-inflammatory drugs decrease the risk of developing AD have encouraged further studies elucidating the role of inflammation in AD. Nanoparticles have become an important focus of neurotherapeutic research because they are an especially effective form of drug delivery. Here, we investigate the potential protective effect of indomethacin-loaded lipid-core nanocapsules (IndOH-LNCs) against cell damage and neuroinflammation induced by amyloid beta (Aβ)1-42 in AD models. Our results show that IndOH-LNCs attenuated Aβ-induced cell death and were able to block the neuroinflammation triggered by Aβ1-42 in organotypic hippocampal cultures. Additionally, IndOH-LNC treatment was able to increase interleukin-10 release and decrease glial activation and c-jun N-terminal kinase phosphorylation. As a model of Aβ-induced neurotoxicity in vivo, animals received a single intracerebroventricular injection of Aβ1-42 (1 nmol/site), and 1 day after Aβ1-42 infusion, they were administered either free IndOH or IndOH-LNCs (1 mg/kg, intraperitoneally) for 14 days. Only the treatment with IndOH-LNCs significantly attenuated the impairment of this behavior triggered by intracerebroventricular injection of Aβ1-42. Further, treatment with IndOH-LNCs was able to block the decreased synaptophysin levels induced by Aβ1-42 and suppress glial and microglial activation. These findings might be explained by the increase of IndOH concentration in brain tissue attained using drug-loaded lipid-core NCs. All these findings support the idea that blockage of neuroinflammation triggered by Aβ is involved in the neuroprotective effects of IndOH-LNCs. These data provide strong evidence that IndOH-LNC treatment may represent a promising approach for treating AD.


European Journal of Pharmacology | 2008

Selective cytotoxicity of indomethacin and indomethacin ethyl ester-loaded nanocapsules against glioma cell lines: an in vitro study.

Andressa Bernardi; Rudimar Luiz Frozza; Eliézer Jäger; Fabrício Figueiró; Luci Bavaresco; Christianne Gazzana Salbego; Adriana Raffin Pohlmann; Guterres Ss; Ana Maria Oliveira Battastini

Gliomas are the most common and devastating tumors of the central nervous system. Several studies have suggested that nonsteroidal anti-inflammatory drugs (NSAIDs) are promising anticancer agents. Biodegradable nanoparticulate systems have received considerable attention as potential drug delivery vehicles. The aim of this study was to evaluate the effects of indomethacin-loaded nanocapsules and indomethacin ethyl ester-loaded nanocapsules on glioma cell lines. In addition, the effect of these formulations on normal neural tissue was also evaluated. In order to investigate this, glioma cell lines (U138-MG and C6) and hippocampal organotypic cultures were used. The main finding of the present study is that indomethacin-loaded nanocapsules formulation was more potent than a solution of indomethacin in decreasing the viability and cell proliferation of glioma lines. Indomethacin and indomethacin ethyl ester associated together in the same nanocapsule formulation caused a synergic effect decreasing glioma cell proliferation. In addition, when the glioma cells were exposed to 25 microM of indomethacin-loaded nanocapsules or indomethacin ethyl ester-loaded nanocapsules, a necrotic cell death was observed. Interestingly, 5 microM of indomethacin-loaded nanocapsules was able to cause an antiproliferative effect without promoting necrosis in glioma cells. Another important finding was that the cytotoxic effect induced by 25 microM or 50 microM of indomethacin-loaded nanocapsules or indomethacin ethyl ester-loaded nanocapsules, in glioma cells was not observed in the organotypic cultures, indicating selective cytotoxicity of those formulations for tumoral cells. Further investigations using in vivo glioma model should be helpful to confirm the distinct effects of indomethacin-loaded nanocapsules and indomethacin ethyl ester-loaded nanocapsules, in normal versus tumoral cells.


Neuroscience Research | 2009

Conditioned medium from mesenchymal stem cells induces cell death in organotypic cultures of rat hippocampus and aggravates lesion in a model of oxygen and glucose deprivation.

Ana Paula Horn; Rudimar Luiz Frozza; Patrícia Benke Grudzinski; Daniéli Gerhardt; Juliana Bender Hoppe; Alessandra Nejar Bruno; Pedro Cesar Chagastelles; Nance Beyer Nardi; Guido Lenz; Christianne Gazzana Salbego

Cell therapy using bone marrow-derived mesenchymal stem cells (MSC) seems to be a new alternative for the treatment of neurological diseases, including stroke. In order to investigate the response of hippocampal tissue to factors secreted by MSC and if these factors are neuroprotective in a model of oxygen and glucose deprivation (OGD), we used organotypic hippocampal cultures exposed to conditioned medium from bone marrow-derived MSC. Our results suggest that the conditioned medium obtained from these cells aggravates lesion caused by OGD. In addition, the presence of the conditioned medium alone was toxic mainly to cells in the CA1, CA2 and CA3 areas of the hippocampal organotypic culture even in basal conditions. GABA stimulation and NMDA and AMPA receptors antagonists were able to reduce propidium iodide staining, suggesting that the cell death induced by the toxic factors secreted by MSC could involve these receptors.


Neurochemistry International | 2011

Amyloid-β induced toxicity involves ganglioside expression and is sensitive to GM1 neuroprotective action.

Fernando Kreutz; Rudimar Luiz Frozza; Ana Carolina Breier; Valeska Aguiar de Oliveira; Ana Paula Horn; Letícia Ferreira Pettenuzzo; Carlos Alexandre Netto; Christianne Gazzana Salbego; Vera Maria Treis Trindade

The effect of Aβ25-35 peptide, in its fibrillar and non-fibrillar forms, on ganglioside expression in organotypic hippocampal slice cultures was investigated. Gangliosides were endogenously labeled with D-[1-C(14)] galactose and results showed that Aβ25-35 affected ganglioside expression, depending on the peptide aggregation state, that is, fibrillar Aβ25-35 caused an increase in GM3 labeling and a reduction in GD1b labeling, whereas the non-fibrillar form was able to enhance GM1 expression. Interestingly, GM1 exhibited a neuroprotective effect in this organotypic model, since pre-treatment of the hippocampal slices with GM1 10 μM was able to prevent the toxicity triggered by the fibrillar Aβ25-35, when measured by propidium iodide uptake protocol. With the purpose of further investigating a possible mechanism of action, we analyzed the effect of GM1 treatment (1, 6, 12 and 24h) upon the Aβ-induced alterations on GSK3β dephosphorylation/activation state. Results demonstrated an important effect after 24-h incubation, with GM1 preventing the Aβ-induced dephosphorylation (activation) of GSK3β, a signaling pathway involved in apoptosis triggering and neuronal death in models of Alzheimers disease. Taken together, present results provide a new and important support for ganglioside participation in development of Alzheimers disease experimental models and suggest a protective role for GM1 in Aβ-induced toxicity. This may be useful for designing new therapeutic strategies for Alzheimers treatment.


Neurochemistry International | 2010

Protective effects of indomethacin-loaded nanocapsules against oxygen-glucose deprivation in organotypic hippocampal slice cultures: Involvement of neuroinflammation

Andressa Bernardi; Rudimar Luiz Frozza; Ana Paula Horn; Maria M. Campos; João B. Calixto; Christianne Gazzana Salbego; Adriana Raffin Pohlmann; Silvia Stanisçuaski Guterres; Ana Maria Oliveira Battastini

Targeted treatment of diseases of the central nervous system remains problematic due to the complex pathogenesis of these disorders and the difficulty in drug delivery. Here we investigate the neuroprotective effect of indomethacin-loaded nanocapsules (IndOH-NC) in an in vitro model of ischemia. For this purpose we used organotypic hippocampal cultures exposed to oxygen-glucose deprivation (OGD). When the cultures were exposed to 60 min of OGD, 54.5±14.7% of the total area of the hippocampal slices was labeled with propidium iodide. On the other hand, when the cultures were treated with 50 or 100 μM of IndOH-NC the cell death was significantly reduced to 31±7% (P<0.05) and 20±4% (P<0.001), respectively. The treatment with IndOH-NC markedly inhibited the levels of the pro-inflammatory cytokines IL-1β, IL-6 and TNF-α levels even 48 h after OGD. Immunoblotting revealed that treatment with 100 μM of IndOH-NC was able to significantly reduce to the levels of control cultures the levels of ERK1/2 and JNK phosphorylation, as well as iNOS activation. Additionally, IndOH-NC prevented glial activation induced by OGD, as evidenced by a decrease of GFAP immunocontent and Isolectin B(4) reactivity. Our results clearly demonstrate that IndOH-NC might represent a promising pharmaceutical neuroprotective formulation for cerebral ischemia, most probably by inhibiting the inflammatory cascades.


International Journal of Nanomedicine | 2013

The antiproliferative effect of indomethacin-loaded lipid-core nanocapsules in glioma cells is mediated by cell cycle regulation, differentiation, and the inhibition of survival pathways

Andressa Bernardi; Rudimar Luiz Frozza; Juliana Bender Hoppe; Christianne Gazzana Salbego; Adriana Raffin Pohlmann; Ana Maria Oliveira Battastini; Silvia Stanisçuaski Guterres

Despite recent advances in radiotherapy, chemotherapy, and surgical techniques, glioblastoma multiforme (GBM) prognosis remains dismal. There is an urgent need for new therapeutic strategies. Nanoparticles of biodegradable polymers for anticancer drug delivery have attracted intense interest in recent years because they can provide sustained, controlled, and targeted delivery. Here, we investigate the mechanisms involved in the antiproliferative effect of indomethacin-loaded lipid-core nanocapsules (IndOH-LNC) in glioma cells. IndOH-LNC were able to reduce cell viability by inducing apoptotic cell death in C6 and U138-MG glioma cell lines. Interestingly, IndOH-LNC did not affect the viability of primary astrocytes, suggesting that this formulation selectively targeted transformed cells. Mechanistically, IndOH-LNC induced inhibition of cell growth and cell-cycle arrest to be correlated with the inactivation of AKT and β-catenin and the activation of GSK-3β. IndOH-LNC also induced G0/G1 and/or G2/M phase arrest, which was accompanied by a decrease in the levels of cyclin D1, cyclin B1, pRb, and pcdc2 and an increase in the levels of Wee1 CDK inhibitor p21WAF1. Additionally, IndOH-LNC promoted GBM cell differentiation, observed as upregulation of glial fibrillary acidic protein (GFAP) protein and downregulation of nestin and CD133. Taken together, the crosstalk among antiproliferative effects, cell-cycle arrest, apoptosis, and cell differentiation should be considered when tailoring pharmacological interventions aimed at reducing glioma growth by using formulations with multiples targets, such as IndOH-LNC.


Stem Cells and Development | 2011

Mesenchymal Stem Cell-Conditioned Medium Triggers Neuroinflammation and Reactive Species Generation in Organotypic Cultures of Rat Hippocampus

Ana Paula Horn; Andressa Bernardi; Rudimar Luiz Frozza; Patrícia Bencke Grudzinski; Juliana Bender Hoppe; Luiz Fernando de Souza; Pedro Cesar Chagastelles; Angela Terezinha de Souza Wyse; Elena Aida Bernard; Ana Maria Oliveira Battastini; Maria M. Campos; Guido Lenz; Nance Beyer Nardi; Christianne Gazzana Salbego

Cell therapy using bone marrow-derived mesenchymal stem cells (MSCs) seems to be a new alternative for the treatment of neurodegenerative diseases. Despite several promising results with their use, possible side effects are still unknown. In a previous work, we have shown that MSC-conditioned medium is toxic to hippocampal slice cultures and aggravates cell death induced by oxygen and glucose deprivation. In this work, we investigated whether the inflammatory response and/or reactive species formation could be involved in that toxicity. Rat organotypic hippocampal cultures were exposed for 24 h to conditioned medium from MSCs isolated from rat bone marrow. A marked glial activation was observed after exposure of cultures to MSC-conditioned medium, as evidenced by glial fibrillary acid protein (GFAP) and isolectin B(4) increase. Tumor necrosis factor-α and interleukin-6 levels were increased in the culture medium, and 2,7-dihydrodichlorofluorescein diacetate oxidation (indicating reactive species generation) and inducible nitric oxide synthase (iNOS) immunocontent were also higher after exposure of cultures to MSC-conditioned medium. Antioxidants (ascorbic acid and TROLOX(®)), N(ω)-nitro-l-arginine methyl ester hydrochloride, and anti-inflammatory drugs (indomethacin and dexamethasone) reduced cell death in hippocampal organotypic cultures after their exposure to MSC-conditioned medium. The results obtained here suggest that MSC-secreted factors trigger reactive species generation and neuroinflammation in organotypic cultures of hippocampus, introducing a note of caution in the use of these cells for neurological application.

Collaboration


Dive into the Rudimar Luiz Frozza's collaboration.

Top Co-Authors

Avatar

Christianne Gazzana Salbego

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Ana Paula Horn

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Andressa Bernardi

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Juliana Bender Hoppe

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Ana Maria Oliveira Battastini

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Lauren Lúcia Zamin

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Melissa Calegaro Nassif

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Daniéli Gerhardt

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Fabrício Simão

Pontifícia Universidade Católica do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Helena Iturvides Cimarosti

Universidade Federal do Rio Grande do Sul

View shared research outputs
Researchain Logo
Decentralizing Knowledge