Network


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

Hotspot


Dive into the research topics where Camila C. Portugal is active.

Publication


Featured researches published by Camila C. Portugal.


Journal of Neurochemistry | 2009

Glutamate receptors modulate sodium-dependent and calcium-independent vitamin C bidirectional transport in cultured avian retinal cells.

Camila C. Portugal; Vivian Sayuri Miya; Karin da Costa Calaza; Rochelle Alberto Martins Santos; Roberto Paes-de-Carvalho

Vitamin C is transported in the brain by sodium vitamin C co‐transporter 2 (SVCT‐2) for ascorbate and glucose transporters for dehydroascorbate. Here we have studied the expression of SVCT‐2 and the uptake and release of [14C] ascorbate in chick retinal cells. SVCT‐2 immunoreactivity was detected in rat and chick retina, specially in amacrine cells and in cells in the ganglion cell layer. Accordingly, SVCT‐2 was expressed in cultured retinal neurons, but not in glial cells. [14C] ascorbate uptake was saturable and inhibited by sulfinpyrazone or sodium‐free medium, but not by treatments that inhibit dehydroascorbate transport. Glutamate‐stimulated vitamin C release was not inhibited by the glutamate transport inhibitor l‐β‐threo‐benzylaspartate, indicating that vitamin C release was not mediated by glutamate uptake. Also, ascorbate had no effect on [3H] d‐aspartate release, ruling out a glutamate/ascorbate exchange mechanism. 2‐Carboxy‐3‐carboxymethyl‐4‐isopropenylpyrrolidine (Kainate) or NMDA stimulated the release, effects blocked by their respective antagonists 6,7‐initroquinoxaline‐2,3‐dione (DNQX) or (5R,2S)‐(1)‐5‐methyl‐10,11‐dihydro‐5H‐dibenzo[a,d]cyclohepten‐5,10‐imine hydrogen maleate (MK‐801). However, DNQX, but not MK‐801 or 2‐amino‐5‐phosphonopentanoic acid (APV), blocked the stimulation by glutamate. Interestingly, DNQX prevented the stimulation by NMDA, suggesting that the effect of NMDA was mediated by glutamate release and stimulation of non‐NMDA receptors. The effect of glutamate was neither dependent on external calcium nor inhibited by 1,2‐bis (2‐aminophenoxy) ethane‐N′,N′,N′,N′,‐tetraacetic acid tetrakis (acetoxy‐methyl ester) (BAPTA‐AM), an internal calcium chelator, but was inhibited by sulfinpyrazone or by the absence of sodium. In conclusion, retinal cells take up and release vitamin C, probably through SVCT‐2, and the release can be stimulated by NMDA or non‐NMDA glutamate receptors.


Neurochemistry International | 2005

Activation of glutamate receptors promotes a calcium-dependent and transporter-mediated release of purines in cultured avian retinal cells: possible involvement of calcium/calmodulin-dependent protein kinase II.

Roberto Paes-de-Carvalho; Bruno V. Dias; Rochele A. Martins; Mariana Rodrigues Pereira; Camila C. Portugal; Claudia Lanfredi

Calcium-dependent release of purines was previously demonstrated in cultures of chick retinal cells stimulated with high potassium concentrations but there is no evidence for an exocytotic mechanism of adenosine release from presynaptic terminals. Here we show that activation of NMDA or alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/kainate glutamate ionotropic receptors promotes a two- to three-fold increase in the release of purines from these cultures. Approximately 96% of intracellular radioactivity is found as nucleotides after incubation with [(3)H]adenosine, but more than 85% of glutamate-stimulated released material is found as inosine (60%), hypoxanthine (19.9%) and adenosine (7.8%). The release is prevented by removal of extracellular calcium, by the transporter blocker nitrobenzylthioinosine, or inhibitors of calcium/calmodulin-dependent protein kinase II (CAMK II). The uptake of [(3)H]adenosine, but not of [(3)H]GABA or [(3)H]choline, is also blocked by 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine (KN62), N-[2-(N-(4-chlorocinnamyl)-N-methylaminomethyl)phenyl-N-[2-hydroxiethyl]-4-methoxybenzenesulfonamide (KN93) or the myristoylated autocamtide-2-related inhibitory peptide, suggesting that the enzyme modulates the nucleoside transporter. The distribution of intracellular purines was not affected by KN62. These results indicate that activation of glutamate receptors triggers the release of purines from retinal cells by a mechanism involving calcium influx, CAMK II and the nitrobenzylthioinosine-sensitive nucleoside transporter. The regulation of adenosine release by glutamate receptors and CAMK II could have important consequences in the presynaptic control of glutamate release.


Free Radical Biology and Medicine | 2015

c-Src deactivation by the polyphenol 3-O-caffeoylquinic acid abrogates reactive oxygen species-mediated glutamate release from microglia and neuronal excitotoxicity

Renato Socodato; Camila C. Portugal; Teresa Canedo; Ivan Domith; Nádia A. de Oliveira; Roberto Paes-de-Carvalho; João B. Relvas; Marcelo Cossenza

3-O-caffeoylquinic acid (3-CQA) is an isomer of chlorogenic acid, which has been shown to regulate lipopolysaccharide-induced tumor necrosis factor production in microglia. Whereas overactivation of microglia is associated with neuronal loss in brain diseases via reactive oxygen species (ROS) production and glutamate excitotoxicity, naïve (nonactivated) microglia are believed to generate little ROS under basal conditions, contributing to the modulation of synaptic activity and nerve tissue repair. However, the signaling pathways controlling basal ROS homeostasis in microglial cells are still poorly understood. Here we used time-lapse microscopy coupled with highly sensitive FRET biosensors (for detecting c-Src activation, ROS generation, and glutamate release) and lentivirus-mediated shRNA delivery to study the pathways involved in antioxidant-regulated ROS generation and how this associates with microglia-induced neuronal cell death. We report that 3-CQA abrogates the acquisition of an amoeboid morphology in microglia triggered by Aβ oligomers or the HIV Tat peptide. Moreover, 3-CQA deactivates c-Src tyrosine kinase and abrogates c-Src activation during proinflammatory microglia stimulation, which shuts off ROS production in these cells. Moreover, forced increment of c-Src catalytic activity by overexpressing an inducible c-Src heteromerization construct in microglia increases ROS production, abrogating the 3-CQA effects. Whereas oxidant (hydrogen peroxide) stimulation dramatically enhances glutamate release from microglia, such release is diminished by the 3-CQA inhibition of c-Src/ROS generation, significantly alleviating cell death in cultures from embryonic neurons. Overall, we provide further mechanistic insight into the modulation of ROS production in cortical microglia, indicating antioxidant-regulated c-Src function as a pathway for controlling microglia-triggered oxidative damage.


Cell Death & Differentiation | 2014

The nitric oxide-cGKII system relays death and survival signals during embryonic retinal development via AKT-induced CREB1 activation

Renato Socodato; Rafael Brito; Camila C. Portugal; N A de Oliveira; K C Calaza; Roberto Paes-de-Carvalho

During early neurogenesis, retinal neuronal cells display a conserved differentiation program in vertebrates. Previous studies established that nitric oxide (NO) and cGMP accumulation regulate essential events in retinal physiology. Here we used pharmacological and genetic loss-of-function to investigate the effects of NO and its downstream signaling pathway in the survival of developing avian retinal neurons in vitro and in vivo. Six-day-old (E6) chick retinal cells displayed increased calcium influx and produced higher amounts of NO when compared with E8 cells. L-arginine (substrate for NO biosynthesis) and S-nitroso-N-acetyl-D,L-penicillamine (SNAP; a nitrosothiol NO donor) promoted extensive cell death in E6 retinas, whereas in E8 both substances decreased apoptosis. The effect of NO at both periods was mediated by soluble guanylyl cyclase (sGC) and cGMP-dependent kinase (cGK) activation. In addition, shRNA-mediated cGKII knockdown prevented NO-induced cell death (E6) and cell survival (E8). This, NO-induced cell death or cell survival was not correlated with an early inhibition of retinal cell proliferation. E6 cells also responded differentially from E8 neurons regarding cyclic AMP-responsive element-binding protein (CREB) activation in the retina in vivo. NO strongly decreased nuclear phospho-CREB staining in E6 but it robustly enhanced CREB phosphorylation in the nuclei of E8 neurons, an effect that was completely abrogated by cGKII shRNAs at both embryonic stages. The ability of NO in regulating CREB differentially during retinal development relied on the capacity of cGKII in decreasing (E6) or increasing (E8) nuclear AKT (V-Akt murine thymoma viral oncogene) activation. Accordingly, inhibiting AKT prevented both cGKII shRNA-mediated CREB upregulation in E6 and SNAP-induced CREB activation in E8. Furthermore, shRNA-mediated in vivo cGKII or in vitro CREB1 knockdown confirmed that NO/cGKII dualistically regulated the downstream CREB1 pathway and caspase activation in the chick retina to modulate neuronal viability. These data demonstrate that NO-mediated cGKII signaling may function to control the viability of neuronal cells during early retinal development via AKT/CREB1 activity.


Glia | 2015

c‐Src function is necessary and sufficient for triggering microglial cell activation

Renato Socodato; Camila C. Portugal; Ivan Domith; Nádia A. de Oliveira; Vivian S. M. Coreixas; Erick Correia Loiola; Tânia Martins; Ana Raquel Santiago; Roberto Paes-de-Carvalho; António F. Ambrósio; João B. Relvas

Microglial cells are the resident macrophages of the central nervous system. Their function is essential for neuronal tissue homeostasis. After inflammatory stimuli, microglial cells become activated changing from a resting and highly ramified cell shape to an amoeboid‐like morphology. These morphological changes are associated with the release of proinflammatory cytokines and glutamate, as well as with high phagocytic activity. The acquisition of such phenotype has been associated with activation of cytoplasmic tyrosine kinases, including those of the Src family (SFKs). In this study, using both in vivo and in vitro inflammation models coupled to FRET‐based time‐lapse microscopy, lentiviruses‐mediated shRNA delivery and genetic gain‐of‐function experiments, we demonstrate that among SFKs c‐Src function is necessary and sufficient for triggering microglia proinflammatory signature, glutamate release, microglia‐induced neuronal loss, and phagocytosis. c‐Src inhibition in retinal neuroinflammation experimental paradigms consisting of intravitreal injection of LPS or ischemia–reperfusion injury significantly reduced microglia activation changing their morphology to a more resting phenotype and prevented neuronal apoptosis. Our data demonstrate an essential role for c‐Src in microglial cell activation. GLIA 2015;63:497–511


Science Signaling | 2017

Caveolin-1–mediated internalization of the vitamin C transporter SVCT2 in microglia triggers an inflammatory phenotype

Camila C. Portugal; Renato Socodato; Teresa Canedo; C. Silva; Tânia Martins; Vivian S. M. Coreixas; Erick Correia Loiola; Burkhard Gess; Dominik Röhr; Ana Raquel Santiago; Peter Young; Richard D. Minshall; Roberto Paes-de-Carvalho; António F. Ambrósio; João B. Relvas

Internalization and degradation of a vitamin C transporter trigger activation of microglia. Vitamin C prevents microglia activation Changes in the abundance of ascorbate, the reduced form of vitamin C, in the central nervous system (CNS) alter neuronal function and are associated with neurodevelopmental and neurodegenerative disorders. Activation of microglia, which occurs in response to tissue damage or pathogens, also contributes to neurodegenerative disease. Portugal et al. showed that the plasma membrane sodium–vitamin C cotransporter 2 (SVCT2) was required for microglia homeostasis in the CNS. Decreasing the amount of SVCT2 in the plasma membrane reduced vitamin C uptake and triggered activation of both primary rodent and human microglia. Treating microglia with ascorbate or preventing the internalization of SVCT2 blocked activation of microglia. These results demonstrate that ascorbate plays an essential role in microglial homeostasis and may prevent the microglial activation that contributes to neurodegenerative disease. Vitamin C is essential for the development and function of the central nervous system (CNS). The plasma membrane sodium–vitamin C cotransporter 2 (SVCT2) is the primary mediator of vitamin C uptake in neurons. SVCT2 specifically transports ascorbate, the reduced form of vitamin C, which acts as a reducing agent. We demonstrated that ascorbate uptake through SVCT2 was critical for the homeostasis of microglia, the resident myeloid cells of the CNS that are essential for proper functioning of the nervous tissue. We found that depletion of SVCT2 from the plasma membrane triggered a proinflammatory phenotype in microglia and resulted in microglia activation. Src-mediated phosphorylation of caveolin-1 on Tyr14 in microglia induced the internalization of SVCT2. Ascorbate treatment, SVCT2 overexpression, or blocking SVCT2 internalization prevented the activation of microglia. Overall, our work demonstrates the importance of the ascorbate transport system for microglial homeostasis and hints that dysregulation of ascorbate transport might play a role in neurological disorders.


Scientific Reports | 2017

Dopamine promotes NMDA receptor hypofunction in the retina through D1 receptor-mediated Csk activation, Src inhibition and decrease of GluN2B phosphorylation.

Renato Socodato; Felipe N. Santiago; Camila C. Portugal; Ivan Domith; Thaísa G. Encarnação; Erick Correia Loiola; Ana Lucia Marques Ventura; Marcelo Cossenza; João B. Relvas; Newton G. Castro; Roberto Paes-de-Carvalho

Dopamine and glutamate are critical neurotransmitters involved in light-induced synaptic activity in the retina. In brain neurons, dopamine D1 receptors (D1Rs) and the cytosolic protein tyrosine kinase Src can, independently, modulate the behavior of NMDA-type glutamate receptors (NMDARs). Here we studied the interplay between D1Rs, Src and NMDARs in retinal neurons. We reveal that dopamine-mediated D1R stimulation provoked NMDAR hypofunction in retinal neurons by attenuating NMDA-gated currents, by preventing NMDA-elicited calcium mobilization and by decreasing the phosphorylation of NMDAR subunit GluN2B. This dopamine effect was dependent on upregulation of the canonical D1R/adenylyl cyclase/cAMP/PKA pathway, of PKA-induced activation of C-terminal Src kinase (Csk) and of Src inhibition. Accordingly, knocking down Csk or overexpressing a Csk phosphoresistant Src mutant abrogated the dopamine-induced NMDAR hypofunction. Overall, the interplay between dopamine and NMDAR hypofunction, through the D1R/Csk/Src/GluN2B pathway, might impact on light-regulated synaptic activity in retinal neurons.


Cellular and Molecular Life Sciences | 2016

Methylphenidate-triggered ROS generation promotes caveolae-mediated transcytosis via Rac1 signaling and c-Src-dependent caveolin-1 phosphorylation in human brain endothelial cells.

Vanessa Coelho-Santos; Renato Socodato; Camila C. Portugal; R. Leitão; Manuel Rito; Marcos Barbosa; Pierre Olivier Couraud; Ignacio A. Romero; Babette B. Weksler; Richard D. Minshall; Carlos Fontes-Ribeiro; Teresa Summavielle; João B. Relvas; Ana P. Silva

Methylphenidate (MPH) is an amphetamine-like stimulant commonly prescribed for attention deficit hyperactivity disorder. Despite its widespread use, the cellular/molecular effects of MPH remain elusive. Here, we report a novel direct role of MPH on the regulation of macromolecular flux through human brain endothelial cells (ECs). MPH significantly increased caveolae-mediated transcytosis of horseradish peroxidase through ECs without affecting paracellular permeability. Using FRET-based live cell imaging, together with pharmacological inhibitors and lentiviral-mediated shRNA knockdown, we demonstrate that MPH promoted ROS generation via activation of Rac1-dependent NADPH oxidase (NOX) and c-Src activation at the plasma membrane. c-Src in turn was shown to mediate the phosphorylation of caveolin-1 (Cav1) on Tyr14 leading to enhanced caveolae formation and transendothelial transport. Accordingly, the inhibition of Cav1 phosphorylation by overexpression of a phosphodefective Cav1Y14F mutant or knocking down Cav1 expression abrogated MPH-induced transcytosis. In addition, both vitamin C and inhibition of NOX blocked MPH-triggered vesicular transport. This study, therefore, identifies Rac1/NOX/c-Src-dependent signaling in MPH-induced increase in transendothelial permeability of brain endothelial cell monolayers via caveolae-mediated transcytosis.


Analytical Methods | 2016

Determination of ascorbic acid in the retina during chicken embryo development using high performance liquid chromatography and UV detection

Débora R. S. Lima; Marcelo Cossenza; Carlos Gustavo Garcia; Camila C. Portugal; Flávia Ferreira de Carvalho Marques; Roberto Paes-de-Carvalho; Annibal Duarte Pereira Netto

The retina is a specialized tissue of the central nervous system (CNS) and it is the only part of the CNS that can be visualized non-invasively. During vertebrate development, the retina originates together with the optic nerve as outgrowths of the developing brain, and in this respect, the avian retina is a very convenient model for neurochemical studies of the CNS. In this study, a HPLC-UV method was developed and validated for the determination of ascorbic acid (AA) in the chicken embryo retina. AA has an important role in the retina because of its antioxidant properties. The developed method showed very good figures of merit (recovery = 91 ± 2%; repeatability and intermediate precision better than 1.67% and 2.53% and a limit of quantification of 0.03 mg L−1). Retinas of two embryo ages (12 days and 18 days) showed AA concentrations of 0.0107 ± 0.0010 and 0.0055 ± 0.0005 μg of AA per μg of protein, respectively, and the statistical comparison of results confirmed the decrease of the AA level. These results seem to correlate well with oxidative stress protection, but this fact is still under investigation. As far as we are aware, this is the first study that demonstrates the HPLC-UV determination of ascorbic acid in the chicken embryo retina and its variation along embryo development.


Toxicology Letters | 2018

Microglia and alcohol meet at the crossroads: Microglia as critical modulators of alcohol neurotoxicity

Joana Henriques; Camila C. Portugal; Teresa Canedo; João B. Relvas; Teresa Summavielle; Renato Socodato

Alcohol use disorders affect millions of people worldwide causing huge social and economic burden on modern society. Excessive alcohol consumption or intoxication provokes severe damage to the body inducing immune suppression, liver damage and neurological disorder. In the central nervous system (CNS), alcohol exposure can lead to neuronal loss, cognitive decline, motor dysfunction, inflammation and impairment of neuroimmune responses. Glial cells, from which microglia represent roughly 10-15%, are primary modulators of the neuroimmune responses and inflammation in the CNS. Here we overview literature relating alcohol exposure with microglia activation and brain inflammation, highlighting that microglia are critical regulators of alcohol responses in the CNS. Different studies indicate that alcohol intake alters the microglial activation spectrum, with the microglial response varying according to the dose, duration, and pattern of alcohol administration. Presently, further investigation is required to establish whether microglia dysfunction initiates or simply amplifies the neurotoxicity of alcohol in the brain. Such knowledge can be greatly facilitated by the use of microglia-specific genetic targeting in animal models and will be critical for the development of better therapeutics for mitigating the neurotoxicity induced by alcohol.

Collaboration


Dive into the Camila C. Portugal's collaboration.

Top Co-Authors

Avatar

Renato Socodato

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivan Domith

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar

Marcelo Cossenza

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Teresa Canedo

Instituto de Biologia Molecular e Celular

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joana Henriques

Instituto de Biologia Molecular e Celular

View shared research outputs
Researchain Logo
Decentralizing Knowledge