Network


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

Hotspot


Dive into the research topics where Paula Pierozan is active.

Publication


Featured researches published by Paula Pierozan.


Critical Care | 2006

Neutrophil apoptosis: a marker of disease severity in sepsis and sepsis-induced acute respiratory distress syndrome

Léa Fialkow; Luciano Fochesatto Filho; Mary Clarisse Bozzetti; Adriana Rosa Milani; Edison Moraes Rodrigues Filho; Roberta Maboni Ladniuk; Paula Pierozan; Rafaela Moraes de Moura; João Carlos Prolla; Eric Vachon; Gregory P. Downey

IntroductionApoptosis of neutrophils (polymorphonuclear neutrophils [PMNs]) may limit inflammatory injury in sepsis and acute respiratory distress syndrome (ARDS), but the relationship between the severity of sepsis and extent of PMN apoptosis and the effect of superimposed ARDS is unknown. The objective of this study was to correlate neutrophil apoptosis with the severity of sepsis and sepsis-induced ARDS.MethodsA prospective cohort study was conducted in intensive care units of three tertiary hospitals in Porto Alegre, southern Brazil. Fifty-seven patients with sepsis (uncomplicated sepsis, septic shock, and sepsis-induced ARDS) and 64 controls were enrolled. Venous peripheral blood was collected from patients with sepsis within 24 hours of diagnosis. All surgical groups, including controls, had their blood drawn 24 hours after surgery. Control patients on mechanical ventilation had blood collected within 24 hours of initiation of mechanical ventilation. Healthy controls were blood donors. Neutrophils were isolated, and incubated ex vivo, and apoptosis was determined by light microscopy on cytospun preparations. The differences among groups were assessed by analysis of variance with Tukeys.ResultsIn medical patients, the mean percentage of neutrophil apoptosis (± standard error of the mean [SEM]) was lower in sepsis-induced ARDS (28% ± 3.3%; n = 9) when compared with uncomplicated sepsis (57% ± 3.2%; n = 8; p < 0.001), mechanical ventilation without infection, sepsis, or ARDS (53% ± 3.0%; n = 11; p < 0.001) and healthy controls (69% ± 1.1%; n = 33; p < 0.001) but did not differ from septic shock (38% ± 3.7%; n = 12; p = 0.13). In surgical patients with sepsis, the percentage of neutrophil apoptosis was lower for all groups when compared with surgical controls (52% ± 3.6%; n = 11; p < 0.001).ConclusionIn medical patients with sepsis, neutrophil apoptosis is inversely proportional to the severity of sepsis and thus may be a marker of the severity of sepsis in this population.


Free Radical Biology and Medicine | 2013

Roundup disrupts male reproductive functions by triggering calcium-mediated cell death in rat testis and Sertoli cells

Vera Lúcia de Liz Oliveira Cavalli; Daiane Cattani; Carla Elise Heinz Rieg; Paula Pierozan; Leila Zanatta; Eduardo Benedetti Parisotto; Danilo Wilhelm Filho; Fátima Regina Mena Barreto Silva; Regina Pessoa-Pureur; Ariane Zamoner

Glyphosate is the primary active constituent of the commercial pesticide Roundup. The present results show that acute Roundup exposure at low doses (36 ppm, 0.036 g/L) for 30 min induces oxidative stress and activates multiple stress-response pathways leading to Sertoli cell death in prepubertal rat testis. The pesticide increased intracellular Ca(2+) concentration by opening L-type voltage-dependent Ca(2+) channels as well as endoplasmic reticulum IP3 and ryanodine receptors, leading to Ca(2+) overload within the cells, which set off oxidative stress and necrotic cell death. Similarly, 30 min incubation of testis with glyphosate alone (36 ppm) also increased (45)Ca(2+) uptake. These events were prevented by the antioxidants Trolox and ascorbic acid. Activated protein kinase C, phosphatidylinositol 3-kinase, and the mitogen-activated protein kinases such as ERK1/2 and p38MAPK play a role in eliciting Ca(2+) influx and cell death. Roundup decreased the levels of reduced glutathione (GSH) and increased the amounts of thiobarbituric acid-reactive species (TBARS) and protein carbonyls. Also, exposure to glyphosate-Roundup stimulated the activity of glutathione peroxidase, glutathione reductase, glutathione S-transferase, γ-glutamyltransferase, catalase, superoxide dismutase, and glucose-6-phosphate dehydrogenase, supporting downregulated GSH levels. Glyphosate has been described as an endocrine disruptor affecting the male reproductive system; however, the molecular basis of its toxicity remains to be clarified. We propose that Roundup toxicity, implicated in Ca(2+) overload, cell signaling misregulation, stress response of the endoplasmic reticulum, and/or depleted antioxidant defenses, could contribute to Sertoli cell disruption in spermatogenesis that could have an impact on male fertility.


Experimental Neurology | 2010

Acute intrastriatal administration of quinolinic acid provokes hyperphosphorylation of cytoskeletal intermediate filament proteins in astrocytes and neurons of rats.

Paula Pierozan; Ariane Zamoner; Ângela Krombauer Soska; Roberta Bristot Silvestrin; Samanta Oliveira Loureiro; Luana Heimfarth; Tadeu Mello e Souza; Moacir Wajner; Regina Pessoa-Pureur

In the present study we investigated the effect of in vivo intrastriatal injection of quinolinic acid (QA) on cytoskeletal proteins in astrocytes and neurons of young rats at early stage (30 min) after infusion. QA (150 nmoles/0.5 microL) significantly increased the in vitro phosphorylation of the low molecular weight neurofilament subunit (NFL) and the glial fibrillary acidic protein (GFAP) of neurons and astrocytes, respectively. This effect was mediated by cAMP-dependent protein kinase A (PKA), protein kinase C (PKC) and Ca(2+)/calmodulin-dependent protein kinase II (PKCaMII). In contrast, mitogen activated protein kinases were not activated by QA infusion. Furthermore, the specific N-methyl-D-aspartate (NMDA) antagonist MK-801 (0.25 mg/kg i.p), the antioxidant L-NAME (60 mg\kg\day), and diphenyldisselenide (PheSe)(2) (0.625 mg\kg\day) injected prior to QA infusion totally prevented QA-induced cytoskeletal hyperphosphorylation. We also observed that QA-induced hyperphosphorylation was targeted at the Ser55 phosphorylating site on NFL head domain, described as a regulatory site for NF assembly in vivo. This effect was fully prevented by MK801, by the PKA inhibitor H89 and by (PheSe)(2), whereas staurosporine (PKC inhibitor) only partially prevented Ser55 phosphorylation. The PKCaMII inhibitor (KN93) and the antioxidant L-NAME failed to prevent the hyperphosphorylation of Ser55 by QA infusion. Therefore, we presume that QA-elicited hyperphosphorylation of the neural cytoskeleton, and specially of NFLSer55, achieved by intrastriatal QA injection could represent an early step in the pathophysiological cascade of deleterious events exerted by QA in rat striatum. Our observations also indicate that NMDA-mediated Ca(2+) events and oxidative stress may be related to the altered protein cytoskeleton hyperphosphorylation observed with important implications for brain function.


Biochimica et Biophysica Acta | 2012

1α,25-Dihydroxyvitamin D3 mechanism of action: Modulation of L-type calcium channels leading to calcium uptake and intermediate filament phosphorylation in cerebral cortex of young rats

Leila Zanatta; Paola Bez Goulart; Renata Gonçalves; Paula Pierozan; Elisa C. Winkelmann-Duarte; Viviane Mara Woehl; Regina Pessoa-Pureur; Fátima Regina Mena Barreto Silva; Ariane Zamoner

The involvement of calcium-mediated signaling pathways in the mechanism of action of 1α,25-dihydroxyvitamin D(3) (1,25D) is currently demonstrated. In this study we found that 1,25D induces nongenomic effects mediated by membrane vitamin D receptor (VDRm) by modulating intermediate filament (IF) phosphorylation and calcium uptake through L-type voltage-dependent calcium channels (L-VDCC) in cerebral cortex of 10 day-old rats. Results showed that the mechanism of action of 1,25D involves intra- and extracellular calcium levels, as well as the modulation of chloride and potassium channels. The effects of L-VDCCs on membrane voltage occur over a broad potential range and could involve depolarizing or hyperpolarizing coupling modes, supporting a cross-talk among Ca(2+) uptake and potassium and chloride channels. Also, the Na(+)/K(+)-ATPase inactivation by ouabain mimicked the 1,25D action on (45)Ca(2+) uptake. The Na(+)/K(+)-ATPase inhibition observed herein might lead to intracellular Na(+) accumulation with subsequent L-VDCC opening and consequently increased (45)Ca(2+) (calcium, isotope of mass 45) uptake. Moreover, the 1,25D effect is dependent on the activation of the following protein kinases: cAMP-dependent protein kinase (PKA), Ca(2+)/calmodulin-dependent protein kinase (PKCaMII), phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase p38 (p38(MAPK)). The modulation of calcium entry into neural cells by the 1,25D we are highlighting, might take a role in the regulation of a plethora of intracellular processes. Considering that vitamin D deficiency can lead to brain illness, 1,25D may be a possible candidate to be used, at least as an adjuvant, in the pharmacological therapy of neuropathological conditions.


Molecular and Cellular Endocrinology | 2013

Congenital hypothyroidism alters the oxidative status, enzyme activities and morphological parameters in the hippocampus of developing rats.

Daiane Cattani; Paola Bez Goulart; Vera Lúcia de Liz Oliveira Cavalli; Elisa C. Winkelmann-Duarte; André Quincozes dos Santos; Paula Pierozan; Daniela Fraga de Souza; Viviane Mara Woehl; Marilda da Cruz Fernandes; Fátima Regina Mena Barreto Silva; Carlos Alberto Saraiva Goncalves; Regina Pessoa-Pureur; Ariane Zamoner

Congenital hypothyroidism is associated with delay in cell migration and proliferation in brain tissue, impairment of synapse formation, misregulation of neurotransmitters, hypomyelination and mental retardation. However, the mechanisms underlying the neuropsychological deficits observed in congenital hypothyroidism are not completely understood. In the present study we proposed a mechanism by which hypothyroidism leads to hippocampal neurotoxicity. Congenital hypothyroidism induces c-Jun-N-terminal kinase (JNK) pathway activation leading to hyperphosphorylation of the glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits from hippocampal astrocytes and neurons, respectively. Moreover, hyperphosphorylation of the cytoskeletal proteins was not reversed by T3 and poorly reversed by T4. In addition, congenital hypothyroidism is associated with downregulation of astrocyte glutamate transporters (GLAST and GLT-1) leading to decreased glutamate uptake and subsequent influx of Ca(2+) through N-methyl-D-aspartate (NMDA) receptors. The Na(+)-coupled (14)C-α-methyl-amino-isobutyric acid ((14)C-MeAIB) accumulation into hippocampal cells also might cause an increase in the intracellular Ca(2+) concentration by opening voltage-dependent calcium channels (VDCC). The excessive influx of Ca(2+) through NMDA receptors and VDCCs might lead to an overload of Ca(2+) within the cells, which set off glutamate excitotoxicity and oxidative stress. The inhibited acetylcholinesterase (AChE) activity might also induce Ca(2+) influx. The inhibited glucose-6-phosphate dehydrogenase (G6PD) and gamma-glutamyl transferase (GGT) activities, associated with altered glutamate and neutral amino acids uptake could somehow affect the GSH turnover, the antioxidant defense system, as well as the glutamate-glutamine cycle. Reduced levels of S100B and glial fibrillary acidic protein (GFAP) take part of the hypothyroid condition, suggesting a compromised astroglial/neuronal neurometabolic coupling which is probably related to the neurotoxic damage in hypothyroid brain.


Experimental Neurology | 2012

Signaling mechanisms downstream of quinolinic acid targeting the cytoskeleton of rat striatal neurons and astrocytes

Paula Pierozan; Ariane Zamoner; Ângela Krombauer Soska; Bárbara Ortiz de Lima; Karina Pires Reis; Fernanda Zamboni; Moacir Wajner; Regina Pessoa-Pureur

The studies of signaling mechanisms involved in the disruption of the cytoskeleton homeostasis were performed in a model of quinolinic acid (QUIN) neurotoxicity in vitro. This investigation focused on the phosphorylation level of intermediate filament (IF) subunits of astrocytes (glial fibrillary acidic protein - GFAP) and neurons (low, medium and high molecular weight neurofilament subunits - NFL, NFM and NFH, respectively). The activity of the phosphorylating system associated with the IFs was investigated in striatal slices of rat exposed to QUIN or treated simultaneously with QUIN plus glutamate receptor antagonists, calcium channel blockers or kinase inhibitors. Results showed that in astrocytes, the action of 100 μM QUIN was mainly due to increased Ca(2+) influx through NMDA and L-type voltage-dependent Ca(2+) channels (L-VDCC). In neuronal cells QUIN acted through metabotropic glutamate receptor (mGluR) activation and influx of Ca(2+) through NMDA receptors and L-VDCC, as well as Ca(2+) release from intracellular stores. These mechanisms then set off a cascade of events including activation of PKA, PKCaMII and PKC, which phosphorylate head domain sites on GFAP and NFL. Also, Cdk5 was activated downstream of mGluR5, phosphorylating the KSP repeats on NFM and NFH. mGluR1 was upstream of phospholipase C (PLC) which, in turn, produced diacylglycerol (DAG) and inositol 3,4,5 triphosphate (IP3). DAG is important to activate PKC and phosphorylate NFL, while IP(3) contributed to Ca(2+) release from internal stores promoting hyperphosphorylation of KSP repeats on the tail domain of NFM and NFH. The present study supports the concept of glutamate and Ca(2+) contribution in excitotoxic neuronal damage provoked by QUIN associated to dysfunction of the cytoskeleton homeostasis and highlights the differential signaling mechanisms elicited in striatal astrocytes and neurons.


Metabolic Brain Disease | 2013

Methylglyoxal-induced cytotoxicity in neonatal rat brain: a role for oxidative stress and MAP kinases

Luana Heimfarth; Samanta Oliveira Loureiro; Paula Pierozan; Bárbara Ortiz de Lima; Karina Pires Reis; Elisandra Barbosa Torres; Regina Pessoa-Pureur

Carbonyl compounds such as methylglyoxal (MGO) seem to play an important role in complications resulting from diabetes mellitus, in aging and neurodegenerative disorders. In this study, we are showing, that MGO is able to suppress cell viability and induce apoptosis in the cerebral cortex and hippocampus of neonatal rats ex-vivo. These effects are partially related with ROS production, evaluated by DCFH-DA assay. Coincubation of MGO and reduced glutathione (GSH) or Trolox (vitamin E) totally prevented ROS production but only partially prevented the MGO-induced decreased cell viability in the two brain structures, as evaluated by the MTT assay. Otherwise, L-NAME, a nitric oxide (NO) inhibitor, partially prevented ROS production in the two structures but partially prevented cytotoxicity in the hippocampus. Pharmacological inhibition of Erk, has totally attenuated MGO-induced ROS production and cytotoxicity, suggesting that MEK/Erk pathway could be upstream of ROS generation and cell survival. Otherwise, p38MAPK and JNK failed to prevent ROS generation but induced decreased cell survival consistent with ROS-independent mechanisms. We can propose that Erk, p38MAPK and JNK are involved in the cytotoxicity induced by MGO through different signaling pathways. While Erk could be an upstream effector of ROS generation, p38MAPK and JNK seem to be associated with ROS-independent cytotoxicity in neonatal rat brain. The cytotoxic damage progressed to apoptotic cell death at MGO concentration higher than those described for adult brain, suggesting that the neonatal brain is resistant to MGO-induced cell death. The consequences of MGO-induced brain damage early in life, remains to be clarified. However, it is feasible that high MGO levels during cortical and hippocampal development could be, at least in part, responsible for the impairment of cognitive functions in adulthood.


Steroids | 2008

Vimentin phosphorylation as a target of cell signaling mechanisms induced by 1α,25-dihydroxyvitamin D3 in immature rat testes

Ariane Zamoner; Paula Pierozan; Luiza Fedatto Vidal; Bruna Arcce Lacerda; Natália Gomes dos Santos; Camila Simioni Vanzin; Regina Pessoa-Pureur

The effects of 1alpha,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] are mainly mediated by nuclear receptors modulating gene expression. However, there are increasing evidences of nongenomic mechanisms of this hormone associated with kinase- and calcium-activated signaling pathways. In this context, the aim of the present work was to investigate the signaling pathways involved in the mechanism of action of 1,25(OH)(2)D(3) on vimentin phosphorylation in 15-day-old rat testes. Results showed that 1,25(OH)(2)D(3) at concentrations ranging from 1 nM to 1 microM increased vimentin phosphorylation independent of protein synthesis. We also demonstrated that the mechanisms underlying the hormone action involve protein kinase C activation in a phospholipase C-independent manner. Moreover, we showed that the participation of protein kinase A, extracellular regulated protein kinase (ERK), and intra- and extracellular Ca(2+) mediating the effects of 1,25(OH)(2)D(3) on the cytoskeleton. In addition, we investigated the effect of different times of exposure to the hormone on total and phosphoERK1/2 or c-Jun N-terminal kinases 1/2 (JNK1/2) in immature rat testis. Results showed that the total levels of ERK1/2 and JNK1/2 were unaltered from 1 to 15 min exposure to 1,25(OH)(2)D(3). However, the phosphoERK1/2 levels significantly increased at 1 and 5 min 1,25(OH)(2)D(3) treatment. Furthermore, phosphoJNK1 levels were decreased at 10 and 15 min 1,25(OH)(2)D(3) exposure, while phosphoJNK 2 levels were diminished at 5, 10 and 15 min treatment with the hormone. These findings demonstrate that 1,25(OH)(2)D(3) may modulate vimentin phosphorylation through nongenomic Ca(2+)-dependent mechanisms in testis cells.


FEBS Journal | 2014

Biochemical, histopathological and behavioral alterations caused by intrastriatal administration of quinolic acid to young rats

Paula Pierozan; Carolina Gonçalves Fernandes; Márcio Ferreira Dutra; Pablo Pandolfo; Fernanda Ferreira; Bárbara Ortiz de Lima; Lisiane O. Porciúncula; Moacir Wajner; Regina Pessoa-Pureur

Quinolinic acid (QUIN) is a neuroactive metabolite of the kinurenine pathway, and is considered to be involved in aging and some neurodegenerative disorders, including Huntingtons disease. QUIN was injected intrastriatally into adolescent rats, and biochemical and histopathological analyses in the striatum, cortex, and hippocampus, as well as behavioral tests, were carried out in the rats over a period of 21 days after drug injection. Decreased [3H]glutamate uptake and increased 45Ca2+ uptake were detected shortly after injection in the striatum and cerebral cortex. In the hippocampus, increased 45Ca2+ uptake preceded the decreased [3H]glutamate uptake, without histopathological alterations. Also, corticostriatal astrogliosis was observed 7 days later, progressing to neuronal death at day 14. QUIN‐treated rats also showed cognitive deficits 24 h after injection, concurrently with striatal astrogliosis. Motor deficits appeared later, after corticostriatal neurodegeneration. We assume that glutamate excitotoxicity could represent, at least in part, a molecular mechanism associated with the cognitive and motor impairments, corticostriatal astrogliosis and neuronal death observed in the QUIN‐treated rats. We propose that our findings could be relevant for understanding the pathophysiology of human neurodegenerative diseases affecting young people, such as the juvenile form of Huntingtons disease, and for the design of potential therapeutic strategies to slow down the progression of the disease.


Journal of Neuroscience Research | 2015

Quinolinic acid induces disrupts cytoskeletal homeostasis in striatal neurons. Protective role of astrocyte–neuron interaction

Paula Pierozan; Fernanda Ferreira; Bárbara Ortiz de Lima; Regina Pessoa-Pureur

Quinolinic acid (QUIN) is an endogenous metabolite of the kynurenine pathway involved in several neurological disorders. Among the several mechanisms involved in QUIN‐mediated toxicity, disruption of the cytoskeleton has been demonstrated in striatally injected rats and in striatal slices. The present work searched for the actions of QUIN in primary striatal neurons. Neurons exposed to 10 µM QUIN presented hyperphosphorylated neurofilament (NF) subunits (NFL, NFM, and NFH). Hyperphosphorylation was abrogated in the presence of protein kinase A and protein kinase C inhibitors H89 (20 μM) and staurosporine (10 nM), respectively, as well as by specific antagonists to N‐methyl‐D‐aspartate (50 µM DL‐AP5) and metabotropic glutamate receptor 1 (100 µM MPEP). Also, intra‐ and extracellular Ca2+ chelators (10 µM BAPTA‐AM and 1 mM EGTA, respectively) and Ca2+ influx through L‐type voltage‐dependent Ca2+ channel (10 µM verapamil) are implicated in QUIN‐mediated effects. Cells immunostained for the neuronal markers βIII‐tubulin and microtubule‐associated protein 2 showed altered neurite/neuron ratios and neurite outgrowth. NF hyperphosphorylation and morphological alterations were totally prevented by conditioned medium from QUIN‐treated astrocytes. Cocultured astrocytes and neurons interacted with one another reciprocally, protecting them against QUIN injury. Cocultured cells preserved their cytoskeletal organization and cell morphology together with unaltered activity of the phosphorylating system associated with the cytoskeleton. This article describes cytoskeletal disruption as one of the most relevant actions of QUIN toxicity in striatal neurons in culture with soluble factors secreted by astrocytes, with neuron–astrocyte interaction playing a role in neuroprotection.

Collaboration


Dive into the Paula Pierozan's collaboration.

Top Co-Authors

Avatar

Regina Pessoa-Pureur

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Angela Terezinha de Souza Wyse

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Luana Heimfarth

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Samanta Oliveira Loureiro

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Felipe Schmitz

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Luiza Fedatto Vidal

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

André Felipe Rodrigues

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Helena Biasibetti

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Ângela Krombauer Soska

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Bruna Arcce Lacerda

Universidade Federal do Rio Grande do Sul

View shared research outputs
Researchain Logo
Decentralizing Knowledge