Network


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

Hotspot


Dive into the research topics where Cláudia S. F. Queiroga is active.

Publication


Featured researches published by Cláudia S. F. Queiroga.


Journal of Biological Chemistry | 2010

Glutathionylation of Adenine Nucleotide Translocase Induced by Carbon Monoxide Prevents Mitochondrial Membrane Permeabilization and Apoptosis

Cláudia S. F. Queiroga; Ana S. Almeida; Cécile Martel; Catherine Brenner; Paula M. Alves; Helena L. A. Vieira

The present work demonstrates the ability of CO to prevent apoptosis in a primary culture of astrocytes. For the first time, the antiapoptotic behavior can be clearly attributed to the inhibition of mitochondrial membrane permeabilization (MMP), a key event in the intrinsic apoptotic pathway. In isolated non-synaptic mitochondria, CO partially inhibits (i) loss of potential, (ii) the opening of a nonspecific pore through the inner membrane, (iii) swelling, and (iv) cytochrome c release, which are induced by calcium, diamide, or atractyloside (a ligand of ANT). CO directly modulates ANT function by enhancing ADP/ATP exchange and prevents its pore-forming activity. Additionally, CO induces reactive oxygen species (ROS) generation, and its prevention by β-carotene decreases CO cytoprotection in intact cells as well as in isolated mitochondria, revealing the key role of ROS. On the other hand, CO induces a slight increase in mitochondrial oxidized glutathione, which is essential for apoptosis modulation by (i) delaying astrocytic apoptosis, (ii) decreasing MMP, and (iii) enhancing ADP/ATP translocation activity of ANT. Moreover, CO and GSSG trigger ANT glutathionylation, a post-translational process regulating protein function in response to redox cellular changes. In conclusion, CO protects astrocytes from apoptosis by preventing MMP, acting on ANT (glutathionylation and inhibition of its pore activity) via a preconditioning-like process mediated by ROS and GSSG.


Journal of Neurochemistry | 2008

Pre‐conditioning induced by carbon monoxide provides neuronal protection against apoptosis

Helena L. A. Vieira; Cláudia S. F. Queiroga; Paula M. Alves

Carbon monoxide (CO) is an endogenous product of mammalian cells generated by heme‐oxygenase, presenting anti‐apoptotic properties in several tissues. The present work demonstrates the ability of small amounts of exogenous CO to prevent neuronal apoptosis induced by excitotoxicity and oxidative stress in mice primary culture of cerebellar granule cells. Additionally, our data show that endogenous CO is a heme‐oxygenase product critical for its anti‐apoptotic activity. Despite being neuroprotective, CO also induces reactive oxygen species generation in neurons. These two phenomena suggest that CO induces pre‐conditioning (PC) to prevent cell death. The role of several PC mediators, namely soluble guanylyl cyclase, nitric oxide (NO) synthase, and ATP‐dependent mitochondrial K channel (mitoKATP) was addressed. Inhibition of soluble guanylyl cyclase or NO synthase activity, or closing of mitoKATP abolishes the protective effect conferred by CO. In addition, CO treatment triggers cGMP and NO production in neurons. Opening of mitoKATP, which appears to be critical for CO prevention of apoptosis, might be a later event. We also demonstrated that reactive oxygen species generation and de novo protein synthesis are necessary for CO PC effect and neuroprotection. In conclusion, CO induces PC and prevents neuronal apoptosis, therefore constituting a novel and promising candidate for neuroprotective therapies.


Journal of Biological Chemistry | 2012

Carbon Monoxide Modulates Apoptosis by Reinforcing Oxidative Metabolism in Astrocytes: ROLE OF Bcl-2*

Ana S. Almeida; Cláudia S. F. Queiroga; Marcos F.Q. Sousa; Paula M. Alves; Helena L. A. Vieira

Background: Low doses of carbon monoxide (CO) prevent apoptosis in several cell models, including astrocytes. Results: CO improves cytochrome c oxidase (COX) activity and induces mitochondrial biogenesis. Bcl-2 expression and interaction with COX is involved in CO signaling. Conclusion: CO stimulates oxidative phosphorylation, improves metabolism, and prevents astrocytic apoptosis. Significance: Metabolism modulation can be a potential strategy against cerebral ischemia. Modulation of cerebral cell metabolism for improving the outcome of hypoxia-ischemia and reperfusion is a strategy yet to be explored. Because carbon monoxide (CO) is known to prevent cerebral cell death; herein the role of CO in the modulation of astrocytic metabolism, in particular, at the level of mitochondria was investigated. Low concentrations of CO partially inhibited oxidative stress-induced apoptosis in astrocytes, by preventing caspase-3 activation, mitochondrial potential depolarization, and plasmatic membrane permeability. CO exposure enhanced intracellular ATP generation, which was accompanied by an increase on specific oxygen consumption, a decrease on lactate production, and a reduction of glucose use, indicating an improvement of oxidative phosphorylation. Accordingly, CO increased cytochrome c oxidase (COX) enzymatic specific activity and stimulated mitochondrial biogenesis. In astrocytes, COX interacts with Bcl-2, which was verified by immunoprecipitation; this interaction is superior after 24 h of CO treatment. Furthermore, CO enhanced Bcl-2 expression in astrocytes. By silencing Bcl-2 expression with siRNA transfection, CO effects in astrocytes were prevented, namely: (i) inhibition of apoptosis, (ii) increase on ATP generation, (iii) stimulation of COX activity, and (iv) mitochondrial biogenesis. Thus, Bcl-2 expression is crucial for CO modulation of oxidative metabolism and for conferring cytoprotection. In conclusion, CO protects astrocytes against oxidative stress-induced apoptosis by improving metabolism functioning, particularly mitochondrial oxidative phosphorylation.


PLOS ONE | 2012

Preconditioning Triggered by Carbon Monoxide (CO) Provides Neuronal Protection Following Perinatal Hypoxia-Ischemia

Cláudia S. F. Queiroga; Simone Tomasi; Marius Widerøe; Paula M. Alves; Alessandro Vercelli; Helena L. A. Vieira

Perinatal hypoxia-ischemia is a major cause of acute mortality in newborns and cognitive and motor impairments in children. Cerebral hypoxia-ischemia leads to excitotoxicity and necrotic and apoptotic cell death, in which mitochondria play a major role. Increased resistance against major damage can be achieved by preconditioning triggered by subtle insults. CO, a toxic molecule that is also generated endogenously, may have a role in preconditioning as low doses can protect against inflammation and apoptosis. In this study, the role of CO-induced preconditioning on neurons was addressed in vitro and in vivo. The effect of 1 h of CO treatment on neuronal death (plasmatic membrane permeabilization and chromatin condensation) and bcl-2 expression was studied in cerebellar granule cells undergoing to glutamate-induced apoptosis. COs role was studied in vivo in the Rice-Vannucci model of neonatal hypoxia-ischemia (common carotid artery ligature +75 min at 8% oxygen). Apoptotic cells, assessed by Nissl staining were counted with a stereological approach and cleaved caspase 3-positive profiles in the hippocampus were assessed. Apoptotic hallmarks were analyzed in hippocampal extracts by Western Blot. CO inhibited excitotoxicity-induced cell death and increased Bcl-2 mRNA in primary cultures of neurons. In vivo, CO prevented hypoxia-ischemia induced apoptosis in the hippocampus, limited cytochrome c released from mitochondria and reduced activation of caspase-3. Still, Bcl-2 protein levels were higher in hippocampus of CO pre-treated rat pups. Our results show that CO preconditioning elicits a molecular cascade that limits neuronal apoptosis. This could represent an innovative therapeutic strategy for high-risk cerebral hypoxia-ischemia patients, in particular neonates.


Biochemistry Research International | 2012

Carbon Monoxide Targeting Mitochondria

Cláudia S. F. Queiroga; Ana S. Almeida; Helena L. A. Vieira

Mitochondria present two key roles on cellular functioning: (i) cell metabolism, being the main cellular source of energy and (ii) modulation of cell death, by mitochondrial membrane permeabilization. Carbon monoxide (CO) is an endogenously produced gaseoustransmitter, which presents several biological functions and is involved in maintaining cell homeostasis and cytoprotection. Herein, mitochondrion is approached as the main cellular target of carbon monoxide (CO). In this paper, two main perspectives concerning CO modulation of mitochondrial functioning are evaluated. First, the role of CO on cellular metabolism, in particular oxidative phosphorylation, is discussed, namely, on: cytochrome c oxidase activity, mitochondrial respiration, oxygen consumption, mitochondrial biogenesis, and general cellular energetic status. Second, the mitochondrial pathways involved in cell death inhibition by CO are assessed, in particular the control of mitochondrial membrane permeabilization.


British Journal of Pharmacology | 2015

Carbon monoxide and the CNS: challenges and achievements

Cláudia S. F. Queiroga; Alessandro Vercelli; Helena L. A. Vieira

Haem oxygenase (HO) and its product carbon monoxide (CO) are associated with cytoprotection and maintenance of homeostasis in several different organs and tissues. This review focuses upon the role of exogenous and endogenous CO (via HO activity and expression) in various CNS pathologies, based upon data from experimental models, as well as from some clinical data on human patients. The pathophysiological conditions reviewed are cerebral ischaemia, chronic neurodegenerative diseases (Alzheimers and Parkinsons diseases), multiple sclerosis and pain. Among these pathophysiological conditions, a variety of cellular mechanisms and processes are considered, namely cytoprotection, cell death, inflammation, cell metabolism, cellular redox responses and vasomodulation, as well as the different targeted neural cells. Finally, novel potential methods and strategies for delivering exogenous CO as a drug are discussed, particularly approaches based upon CO‐releasing molecules, their limitations and challenges. The diagnostic and prognostic value of HO expression in clinical use for brain pathologies is also addressed.


BMC Cell Biology | 2011

Carbon monoxide prevents hepatic mitochondrial membrane permeabilization

Cláudia S. F. Queiroga; Ana S. Almeida; Paula M. Alves; Catherine Brenner; Helena L. A. Vieira

BackgroundLow concentrations of carbon monoxide (CO) protect hepatocytes against apoptosis and confers cytoprotection in several models of liver. Mitochondria are key organelles in cell death control via their membrane permeabilization and the release of pro-apoptotic factors.ResultsHerein, we show that CO prevents mitochondrial membrane permeabilization (MMP) in liver isolated mitochondria. Direct and indirect approaches were used to evaluate MMP inhibition by CO: mitochondrial swelling, mitochondrial depolarization and inner membrane permeabilization. Additionally, CO increases mitochondrial reactive oxygen species (ROS) generation, and their scavenging, by ß-carotene addition, decreases CO protection, which reveals the key role of ROS. Interestingly, cytochrome c oxidase transiently responds to low concentrations of CO by decreasing its activity in the first 5 min, later on there is an increase of cytochrome c oxidase activity, which were detected up to 30 min.ConclusionCO directly prevents mitochondrial membrane permeabilization, which might be implicated in the hepatic apoptosis inhibition by this gaseoustransmitter.


The Journal of Physiology | 2016

Mitochondria and carbon monoxide: cytoprotection and control of cell metabolism – a role for Ca2+?

Sara Oliveira; Cláudia S. F. Queiroga; Helena L. A. Vieira

Carbon monoxide (CO) is an endogenously produced gasotransmitter with important biological functions: anti‐inflammation, anti‐apoptosis, vasomodulation and cell metabolism modulation. The most recognized cellular target for CO is the mitochondria. Physiological concentrations of CO generate mitochondrial reactive oxygen species (ROS), which are signalling molecules for CO‐induced pathways. Indeed, small amounts of ROS promote cytoprotection by a preconditioning effect. Furthermore, CO prevents cell death by limiting mitochondrial membrane permeabilization, which inhibits the release of pro‐apoptotic factors into the cytosol; both events are ROS dependent. CO also increases the ability of mitochondria to take up Ca2+. Mitochondrial metabolism is modulated by CO, namely by increasing TCA cycle rate, oxidative phosphorylation and mitochondrial biogenesis, which, in turn, increases ATP production. COs modulation of metabolism might be important for cellular response to diseases, namely cancer and ischaemic diseases. Finally, another cytoprotective role of CO involves the control of Ca2+ channels. By limiting the activity of T‐type and L‐type Ca2+ channels, CO prevents excitotoxicity‐induced cell death and modulates cell proliferation. Several questions concerning Ca2+ signalling, mitochondria and CO can be asked, for instance whether CO modulation of cell metabolism would be dependent on the mitochondrial Ca2+ uptake capacity, since small amounts of Ca2+ can increase mitochondrial metabolism. Whether CO controls Ca2+ communication between mitochondria and endoplasmic reticulum is another open field of research. In summary, CO emerges as a key gasotransmitter in the control of several cellular functions of mitochondria: metabolism, cell death and Ca2+ signalling.


Journal of Cell Science | 2016

Paracrine effect of carbon monoxide – astrocytes promote neuroprotection through purinergic signaling in mice

Cláudia S. F. Queiroga; Raquel M. A. Alves; Silvia V. Conde; Paula M. Alves; Helena L. A. Vieira

ABSTRACT The neuroprotective role of carbon monoxide (CO) has been studied in a cell-autonomous mode. Herein, a new concept is disclosed – CO affects astrocyte–neuron communication in a paracrine manner to promote neuroprotection. Neuronal survival was assessed when co-cultured with astrocytes that had been pre-treated or not with CO. The CO-pre-treated astrocytes reduced neuronal cell death, and the cellular mechanisms were investigated, focusing on purinergic signaling. CO modulates astrocytic metabolism and extracellular ATP content in the co-culture medium. Moreover, several antagonists of P1 adenosine and P2 ATP receptors partially reverted CO-induced neuroprotection through astrocytes. Likewise, knocking down expression of the neuronal P1 adenosine receptor A2A-R (encoded by Adora2a) reverted the neuroprotective effects of CO-exposed astrocytes. The neuroprotection of CO-treated astrocytes also decreased following prevention of ATP or adenosine release from astrocytic cells and inhibition of extracellular ATP metabolism into adenosine. Finally, the neuronal downstream event involves TrkB (also known as NTRK2) receptors and BDNF. Pharmacological and genetic inhibition of TrkB receptors reverts neuroprotection triggered by CO-treated astrocytes. Furthermore, the neuronal ratio of BDNF to pro-BDNF increased in the presence of CO-treated astrocytes and decreased whenever A2A-R expression was silenced. In summary, CO prevents neuronal cell death in a paracrine manner by targeting astrocytic metabolism through purinergic signaling. Summary: CO modulates astrocytic metabolism and initiates communication pathways in a paracrine manner in order to rescue neurons from cell death, which is dependent on P1 adenosine, P2 ATP and TrkB receptors.


SpringerPlus | 2015

Carbon monoxide targeting mitochondria in astrocytes: modulation of cell metabolism, redox response and cell death.

Helena L. A. Vieira; Ana P.C. Almeida; Cláudia Figueiredo-Pereira; Cláudia S. F. Queiroga

The endogenously produced gasotransmitter carbon monoxide (CO) has been studied as a factor involved in cytoprotection, homeostasis and anti-inflammation. Small amounts of reactive oxygen species (ROS) are described as signaling factors in CO’s biological mode of action. Mitochondria are the main source of ROS and are also key organelles in orchestrating cell function: metabolism, cell death control and redox signaling. Astrocytes are most abundant glial cells and essential for neuronal function, namely metabolic and physical support, expression of neurotransmitters and promotion of neuroprotection. In this work it is shown that CO prevents astrocytic cell death and improves cell metabolism by targeting mitochondria, and some of the underlying molecular mechanism are disclosed. CO directly targets non-synaptic mitochondria and inhibits their mitochondrial membrane permeabilization, by preventing mitochondrial swelling, depolarization and inner membrane permeabilization. Thus, CO limits the release of cytochrome c into the cytosol and the activation of apoptotic cascade in astrocytes. All these events are ROS-dependent and involve glutathionylation of adenine nucleotide translocator (ANT), whose activity is ATP/ADP transport through mitochondrial inner membrane. In addition, low amounts of exogenous CO increase ATP production by improving oxidative metabolism. Mitochondrial population and specific cytochrome c oxidase activity are higher upon CO treatment. The CO-induced metabolic improvement is dependent on Bcl-2 expression. Dysfunctional mitochondrial can be eliminated by mitophagy, which is a crucial process for maintaining their function and quality control. In astrocytes, CO promotes mitophagy at 1h of treatment, while following 24h mitochondrial population is back to basal levels, indicating that CO contributes to mitochondrial turnover. Furthermore, CO limits astrocytic cell death in an autophagic dependent manner.

Collaboration


Dive into the Cláudia S. F. Queiroga's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paula M. Alves

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Ana S. Almeida

Universidade Nova de Lisboa

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana P.C. Almeida

Universidade Nova de Lisboa

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvia V. Conde

Universidade Nova de Lisboa

View shared research outputs
Researchain Logo
Decentralizing Knowledge