Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 59(2): 872-889, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34796462

ABSTRACT

Microglia, the 'resident immunocompetent cells' of the central nervous system (CNS), are key players in innate immunity, synaptic refinement and homeostasis. Dysfunctional microglia contribute heavily to creating a toxic inflammatory milieu, a driving factor in the pathophysiology of several CNS disorders. Therefore, strategies to modulate the microglial function are required to tackle exacerbated tissue inflammation. Carbon monoxide (CO), an endogenous gaseous molecule produced by the degradation of haem, has anti-inflammatory, anti-apoptotic, and pro-homeostatic and cytoprotective roles, among others. ALF-826A, a novel molybdenum-based CO-releasing molecule, was used for the assessment of neuron-microglia remote communication. Primary cultures of rat microglia and neurons, or the BV-2 microglial and CAD neuronal murine cell lines, were used to study the microglia-neuron interaction. An approach based on microglial-derived conditioned media in neuronal culture was applied. Medium derived from CO-treated microglia provided indirect neuroprotection against inflammation by limiting the lipopolysaccharide (LPS)-induced expression of reactivity markers (CD11b), the production of reactive oxygen species (ROS) and the secretion of inflammatory factors (TNF-α, nitrites). This consequently prevented neuronal cell death and maintained neuronal morphology. In contrast, in the absence of inflammatory stimulus, conditioned media from CO-treated microglia improved neuronal morphological complexity, which is an indirect manner of assessing neuronal function. Likewise, the microglial medium also prevented neuronal cell death induced by pro-oxidant tert-Butyl hydroperoxide (t-BHP). ALF-826 treatment reinforced microglia secretion of Interleukin-10 (IL-10) and adenosine, mediators that may protect against t-BHP stress in this remote communication model. Chemical inhibition of the adenosine receptors A2A and A1 reverted the CO-derived neuroprotective effect, further highlighting a role for CO in regulating neuron-microglia communication via purinergic signalling. Our findings indicate that CO has a modulatory role on microglia-to-neuron communication, promoting neuroprotection in a non-cell autonomous manner. CO enhances the microglial release of neurotrophic factors and blocks exacerbated microglial inflammation. CO improvement of microglial neurotrophism under non-inflammatory conditions is here described for the first time.


Subject(s)
Microglia , Neuroprotective Agents , Animals , Carbon Monoxide/metabolism , Carbon Monoxide/pharmacology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats
2.
J Cell Sci ; 129(16): 3178-88, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27383770

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.


Subject(s)
Astrocytes/metabolism , Carbon Monoxide/pharmacology , Neuroprotection/drug effects , Paracrine Communication/drug effects , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , Adenosine/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Coculture Techniques , Cysteine/metabolism , Extracellular Space/metabolism , Gene Silencing/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Mice, Inbred C57BL , Models, Biological , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pyrimidines/pharmacology , Receptor, trkB/metabolism , Receptors, Adenosine A2/metabolism , Serine/metabolism , Suramin/pharmacology , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Triazoles/pharmacology , Xanthines/pharmacology
3.
J Physiol ; 594(15): 4131-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26377343

ABSTRACT

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 Ca(2+) . Mitochondrial metabolism is modulated by CO, namely by increasing TCA cycle rate, oxidative phosphorylation and mitochondrial biogenesis, which, in turn, increases ATP production. CO's 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 Ca(2+) channels. By limiting the activity of T-type and L-type Ca(2+) channels, CO prevents excitotoxicity-induced cell death and modulates cell proliferation. Several questions concerning Ca(2+) signalling, mitochondria and CO can be asked, for instance whether CO modulation of cell metabolism would be dependent on the mitochondrial Ca(2+) uptake capacity, since small amounts of Ca(2+) can increase mitochondrial metabolism. Whether CO controls Ca(2+) 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 Ca(2+) signalling.


Subject(s)
Calcium/metabolism , Carbon Monoxide/metabolism , Mitochondria/metabolism , Animals , Calcium Channels/physiology , Carbon Monoxide/physiology , Cell Death , Cytoprotection , Humans , Reactive Oxygen Species/metabolism
4.
Br J Pharmacol ; 172(6): 1533-45, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24758548

ABSTRACT

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 (Alzheimer's and Parkinson's 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.


Subject(s)
Carbon Monoxide/metabolism , Central Nervous System Diseases/physiopathology , Heme Oxygenase (Decyclizing)/metabolism , Animals , Carbon Monoxide/administration & dosage , Cell Death/physiology , Central Nervous System Diseases/diagnosis , Homeostasis , Humans , Prognosis
5.
PLoS One ; 7(8): e42632, 2012.
Article in English | MEDLINE | ID: mdl-22952602

ABSTRACT

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. CO's 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.


Subject(s)
Carbon Monoxide/chemistry , Hypoxia-Ischemia, Brain , Neurons/metabolism , Animals , Apoptosis , Carotid Artery, Common/pathology , Caspase 3/metabolism , Cells, Cultured , Hippocampus/metabolism , Hippocampus/pathology , Inflammation , Necrosis/pathology , Proto-Oncogene Proteins c-bcl-2 , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors
6.
Biochem Res Int ; 2012: 749845, 2012.
Article in English | MEDLINE | ID: mdl-22536507

ABSTRACT

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.

7.
J Biol Chem ; 287(14): 10761-70, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334654

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Astrocytes/cytology , Astrocytes/metabolism , Carbon Monoxide/pharmacology , Cytoprotection/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Adenosine Triphosphate/metabolism , Animals , Astrocytes/drug effects , Cell Hypoxia/drug effects , Electron Transport Complex IV/metabolism , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Reperfusion Injury/pathology
8.
BMC Cell Biol ; 12: 10, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21388535

ABSTRACT

BACKGROUND: Low 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. RESULTS: Herein, 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. CONCLUSION: CO directly prevents mitochondrial membrane permeabilization, which might be implicated in the hepatic apoptosis inhibition by this gaseoustransmitter.


Subject(s)
Carbon Monoxide/pharmacology , Cytoprotection/drug effects , Mitochondria, Liver , Mitochondrial Membranes , Animals , Apoptosis/drug effects , Electron Transport Complex IV/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Swelling/drug effects , Permeability/drug effects , Reactive Oxygen Species/metabolism , beta Carotene/metabolism
9.
J Biol Chem ; 285(22): 17077-88, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20348099

ABSTRACT

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 beta-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.


Subject(s)
Apoptosis , Carbon Monoxide/chemistry , Glutathione/metabolism , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Membranes/metabolism , Animals , Astrocytes/cytology , Brain/metabolism , Cytochromes c/metabolism , Male , Membrane Potentials , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species
10.
J Neurochem ; 107(2): 375-84, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18691384

ABSTRACT

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 (mitoK(ATP)) was addressed. Inhibition of soluble guanylyl cyclase or NO synthase activity, or closing of mitoK(ATP) abolishes the protective effect conferred by CO. In addition, CO treatment triggers cGMP and NO production in neurons. Opening of mitoK(ATP), 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.


Subject(s)
Apoptosis/drug effects , Carbon Monoxide/metabolism , Carbon Monoxide/pharmacology , Neurons/physiology , Animals , Animals, Newborn , Apoptosis/physiology , Cells, Cultured , Cerebellum/cytology , Decanoic Acids/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Hydroxy Acids/pharmacology , Metalloporphyrins/pharmacology , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/drug effects , Oxadiazoles/pharmacology , Oxidative Stress/physiology , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Time Factors , tert-Butylhydroperoxide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...