Network


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

Hotspot


Dive into the research topics where Karin da Costa Calaza is active.

Publication


Featured researches published by Karin da Costa Calaza.


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.


International Journal of Developmental Neuroscience | 2007

Signal transduction pathways associated with ATP-induced proliferation of cell progenitors in the intact embryonic retina

Patricia Helena Castro Nunes; Karin da Costa Calaza; Lidiane Martins Albuquerque; Lucianne Fragel-Madeira; Alfred Sholl-Franco; Ana Lucia Marques Ventura

ATP and ADP induce retinal cell proliferation through activation of PKC and extracellular signal‐regulated kinases (ERKs). Here, we characterized the effect of purinergic agonists on the turnover of phosphoinositides and activation of ERKs during development of the chick embryo retina. When intact retinas were incubated with ATP, ADP or UTP, a dose‐dependent accumulation of [3H]‐phosphoinositides was observed (% of control, EC50: 548 ± 20.5%, 0.18 mM; 314 ± 53.8%, 0.51 mM; 704 ± 139.9%, 0.018 mM, respectively). Only the response promoted by ADP was completely inhibited by the P2 receptor antagonists, PPADS and suramin. All the responses decreased with the progression of retinal development. Western blot assays revealed that ATP, ADP and UTP stimulated the phosphorylation of ERKs in the chick embryo retina very early during development (% of control: 174 ± 16; 199 ± 16.4 and 206 ± 37, respectively). The responses to ADP and UTP were transient and dose‐dependent, showing EC50 values of 0.12 mM and 0.009 mM. The response to ADP was inhibited by the antagonists PPADS and suramin and by U73122 and chelerythrine chloride, which block PLC and PKC, respectively. Conversely, chelerythrine chloride did not block the response induced by UTP. Immunohistochemical analysis revealed that ATP and ADP induced the phosphorylation of ERKs in cells of the neuroblastic layer of retinas from embryos at E8. Our data showed that ATP, ADP and UTP stimulate the turnover of InsPs and promoted the activation of ERKs in the chick embryo retina. ADP, through activation of P2Y1 receptors, activated ERK pathway through PLC and PKC and UTP, via P2Y4‐like receptors, induced the phosphorylation of ERKs through a pathway that did not involve PKC.


Vision Research | 2009

Modulation of GABA release by nitric oxide in the chick retina: Different effects of nitric oxide depending on the cell population

R.S. Maggesissi; Patrícia F. Gardino; E.M. Guimarães-Souza; R. Paes-de-Carvalho; R.B. Silva; Karin da Costa Calaza

gamma-Aminobutyric acid (GABA) is considered to be the most important inhibitory neurotransmitter in the central nervous system, including the retina. It has been shown that nitric oxide (NO) can influence the physiology of all retinal neuronal types, by mechanisms including modulation of GABA release. However, until now, there have been no data concerning the effects on endogenous GABA release of NO produced by cells in the intact retina. In the present study, we have investigated how NO production induced by drugs influences the release of endogenous GABA in cells of the intact retina of mature chicken. Retinas were exposed to different drugs that affect NO production, and GABA remaining in the tissue was detected by immunohistochemical procedures. A specific nNOS inhibitor (7-NI) reduced the number of GABA+amacrine cells and cells in the ganglion cell layer (GCL) by 33% and 41%, respectively. A GABA transporter inhibitor blocked this effect. L-arginine (100 microM), the precursor of NO, induced increases of 62% and 34% in the number of GABA+amacrine cells and GCL cells, respectively. A sodium (Na(+))-free solution, 7-NI and a PKG inhibitor prevented the effect of L-arginine (100 microM). However, a higher concentration of L-arginine (1mM) induced a 35% reduction in the number of GABA+cells by a Na(+)-dependent mechanism that was restricted to the GCL population. NMDA, which stimulates NO production, increased GABA release as indicated by 53% and 38% reductions in the number of GABA+amacrine cells and GCL cells, respectively. This effect was blocked by 7-NI only in GCL cells. We conclude that basal NO production and moderate NO production (possibly induced by L-arginine; 100 microM) inhibit basal GABA release from amacrine cells and GCL cells. However, NMDA or L-arginine (1mM) induce a NO-dependent increase in GABA release in GCL cells, possibly by stimulating higher NO production.


Journal of Neurochemistry | 2011

Developmental regulation of neuronal survival by adenosine in the in vitro and in vivo avian retina depends on a shift of signaling pathways leading to CREB phosphorylation or dephosphorylation

Renato Socodato; Rafael Brito; Karin da Costa Calaza; Roberto Paes-de-Carvalho

J. Neurochem. (2011) 116, 227–239.


Neurochemical Research | 2003

Local Differences in GABA Release Induced by Excitatory Amino Acids During Retina Development: Selective Activation of NMDA Receptors by Aspartate in the Inner Retina

Karin da Costa Calaza; Maria Christina F. de Mello; Fernando G. de Mello; Patrícia F. Gardino

Glutamate and GABA are the major excitatory and inhibitory neurotransmitters in the CNS. In the retina, it has been shown that glutamate and aspartate and their agonists kainate and NMDA promote the release of GABA. In the chick retina, at embryonic day 14 (E14), glutamate and kainate were able to induce the release of GABA from amacrine and horizontal cells as detected by GABA-immunoreactivity. NMDA also induced GABA release restricted to amacrine cell population and its projections to the inner plexiform layer (E14 and E18). Although aspartate reduced GABA immunoreactivity, specifically in amacrine cells of E18 retinas, it was not efficient to promote GABA release from retinas at E14. As observed in differentiated retinas, dopamine inhibited the GABA release promoted by NMDA and aspartate but not by kainate. Our data show that different retinal sites respond to distinct EAAs via different receptor systems.


Biology Letters | 2015

Near-infrared light increases ATP, extends lifespan and improves mobility in aged Drosophila melanogaster

Rana Begum; Karin da Costa Calaza; Jaimie Hoh Kam; T.E. Salt; Chris Hogg; Glen Jeffery

Ageing is an irreversible cellular decline partly driven by failing mitochondrial integrity. Mitochondria accumulate DNA mutations and reduce ATP production necessary for cellular metabolism. This is associated with inflammation. Near-infrared exposure increases retinal ATP in old mice via cytochrome c oxidase absorption and reduces inflammation. Here, we expose fruitflies daily to 670 nm radiation, revealing elevated ATP and reduced inflammation with age. Critically, there was a significant increase in average lifespan: 100–175% more flies survived into old age following 670 nm exposure and these had significantly improved mobility. This may be a simple route to extending lifespan and improving function in old age.


Journal of Neurochemistry | 2012

Expression of A1 adenosine receptors in the developing avian retina: in vivo modulation by A2A receptors and endogenous adenosine

Rafael Brito; Mariana Rodrigues Pereira; Roberto Paes-de-Carvalho; Karin da Costa Calaza

Little is known about the mechanisms that regulate the expression of adenosine receptors during CNS development. We demonstrate here that retinas from chick embryos injected in ovo with selective adenosine receptor ligands show changes in A1 receptor expression after 48 h. Exposure to A1 agonist N6‐cyclohexyladenosine (CHA) or antagonist 8‐Cyclopentyl‐1, 3‐dipropylxanthine (DPCPX) reduced or increased, respectively, A1 receptor protein and [3H]DPCPX binding, but together, CHA+DPCPX had no effect. Interestingly, treatment with A2A agonist 3‐[4‐[2‐[[6‐amino‐9‐[(2R,3R,4S,5S)‐5‐(ethylcarbamoyl)‐3,4‐dihydroxy‐oxolan‐2‐yl]purin‐2‐yl]amino] ethyl]phenyl] propanoic acid (CGS21680) increased A1 receptor protein and [3H]DPCPX binding, and reduced A2A receptors. The A2A antagonists 7‐(2‐phenylethyl)‐5‐amino‐2‐(2‐furyl)‐pyrazolo‐[4,3‐e]‐1,2,4‐trizolo[1,5‐c] pyrimidine (SCH58261) and 4‐(2‐[7‐amino‐2‐[2‐furyl][1,2,4]triazolo[2,3‐a][1,3,5]triazo‐5‐yl‐amino]ethyl)phenol (ZM241385) had opposite effects on A1 receptor expression. Exposure to CGS21680 + CHA did not change A1 receptor levels, whereas CHA + ZM241385 or CGS21680 + DPCPX had no synergic effect. The blockade of adenosine transporter with S‐(4‐nitrobenzyl)‐6‐thioinosine (NBMPR) also reduced [3H]DPCPX binding, an effect blocked by DPCPX, but not enhanced by ZM241385. [3H]DPCPX binding kinetics showed that treatment with CHA reduced and CGS21680 increased the Bmax, but did not affect Kd values. CHA, DPCPX, CGS21680, and ZM241385 had no effect on A1 receptor mRNA. These data demonstrated an in vivo regulation of A1 receptor expression by endogenous adenosine or long‐term treatment with A1 and A2A receptors modulators.


International Journal of Developmental Neuroscience | 2010

Ethanol increases GABA release in the embryonic avian retina

Fernanda Pohl-Guimarães; Karin da Costa Calaza; Edna N. Yamasaki; Regina Célia Cussa Kubrusly; Ricardo Augusto de Melo Reis

Several mechanisms underlying ethanol action in GABAergic synapses have been proposed, one of these mechanisms is on GABA release. Here, we report that in ovo exposure to ethanol induces an increase on GABA release in the embryonic chick retina. Eleven‐day‐old chick embryos (E11) received an injection of either phosphate buffer saline (PBS) or ethanol (10%, v/v, diluted in PBS), and were allowed to develop until E16. A single glutamate stimulus (2 mM) showed approximately a 40% increase on GABA release in E16 retinas when compared to controls. The effect was dependent on NMDA receptors and GAD65 mRNA levels, which were increased following the ethanol treatment. However, the numbers of GABA‐, GAD‐, and NR1‐immunoreactive cells, and the expression levels of these proteins, were not affected. We conclude that ethanol treatment at a time point when synapses are being formed during development selectively increases GABA release in the retina via a NMDA receptor‐dependent process.


Experimental Brain Research | 2006

GABAergic circuitry in the opossum retina: a GABA release induced by l-aspartate

Karin da Costa Calaza; Jan Nora Hokoç; Patrícia F. Gardino

Glutamate and γ-amino butyric acid (GABA) are the major excitatory and inhibitory neurotransmitters, respectively, in the central nervous system (CNS), including the retina. Although in a number of studies the retinal source of GABA was identified, in several species, as horizontal, amacrine cells and cells in the ganglion cell layer, nothing was described for the opossum retina. Thus, the first goal of this study was to determine the pattern of GABAergic cell expression in the South America opossum retina by using an immunohistochemical approach for GABA and for its synthetic enzyme, glutamic acid decarboxylase (GAD). GABA and GAD immunoreactivity showed a similar cellular pattern by appearing in a few faint horizontal cells, topic and displaced amacrine cells. In an effort to extend the knowledge of the opossum retinal circuitry, the possible influence of glutamatergic inputs in GABAergic cells was also studied. Retinas were stimulated with different glutamatergic agonists and aspartate (Asp), and the GABA remaining in the tissue was detected by immunohistochemical procedures. The exposure of retinas to NMDA and kainate resulted the reduction of the number of GABA immunoreactive topic and displaced amacrine cells. The Asp treatment also resulted in reduction of the number of GABA immunoreactive amacrine cells but, in contrast, the displaced amacrine cells were not affected. Finally, the Asp effect was totally blocked by MK-801. This result suggests that Asp could be indeed a putative neurotransmitter in this non-placental animal by acting on an amacrine cell sub-population of GABA-positive NMDA-sensitive cells.


Journal of Chemical Neuroanatomy | 2004

Identification of neurons with acetilcholinesterase and NADPH-diaphorase activities in the centrifugal visual system of the chick

Patrícia F. Gardino; Alice R Schmal; Karin da Costa Calaza

The isthmo-optic nuclei (ION) and ectopic neurons, which constitute the centrifugal visual system (CVS), are thought to be cholinoceptive and nitrergic. However, it is not clear which neurons express these markers, namely the ones that project to the retina rather than in neurons that only participate in a local circuit. Therefore, to characterize the neurochemical patterns of the centrifugal visual system in the post-hatched chick, retinopetal cells of the isthmo-optic nuclei and the ectopic region were identified via immunolabeling for cholera toxin, a neuronal tracer, which has been injected in the ocular globe. Then, double labeled with acetylcholinesterase histochemistry to reveal cholinergic synapses, or NADPH-diaphorase histochemistry as a nitrergic marker. Briefly, acetylcholinesterase activity was present mainly in cholera toxin labeled cell bodies of the isthmo-optic nucleus and the ectopic region indicating that retinal projecting neurons of centrifugal visual system comprise a cholinoceptive pathway. On the other hand, NADPH-diaphorase histochemistry was present in the neuropile and sparse cell bodies inside of the isthmo-optic nucleus and in ectopic neurons which were not cholera toxin positive suggesting their role in an intrinsic circuit of the centrifugal visual system. These data support the idea that these two neurochemical systems are present in distinct neuronal populations in the centrifugal visual system.

Collaboration


Dive into the Karin da Costa Calaza's collaboration.

Top Co-Authors

Avatar

Patrícia F. Gardino

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcelo Cossenza

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar

E.M. Guimarães-Souza

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fernando G. de Mello

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Ivan Domith

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar

Rafael Brito

Federal Fluminense University

View shared research outputs
Top Co-Authors

Avatar

Ricardo Augusto de Melo Reis

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge