Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Biol Chem ; 299(8): 105036, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37442232

RESUMEN

Arsenic contamination of groundwater is among one of the biggest health threats affecting millions of people in the world. There is an urgent need for efficient arsenic biosensors where the use of arsenic metabolizing enzymes can be explored. In this work, we have solved four crystal structures of arsenite oxidase (Aio) in complex with arsenic and antimony oxyanions and the structures determined correspond to intermediate states of the enzymatic mechanism. These structural data were complemented with density-functional theory calculations providing a unique view of the molybdenum active site at different time points that, together with mutagenesis data, enabled to clarify the enzymatic mechanism and the molecular determinants for the oxidation of As(III) to the less toxic As(V) species.


Asunto(s)
Arsénico , Arsenitos , Humanos , Antimonio , Oxidación-Reducción
2.
Artículo en Inglés | MEDLINE | ID: mdl-31332064

RESUMEN

In the last decade, carbon monoxide-releasing molecules (CORMs) have been shown to act against several pathogens and to be promising antimicrobials. However, the understanding of the mode of action and reactivity of these compounds on bacterial cells is still deficient. In this work, we used a metabolomics approach to probe the toxicity of the ruthenium(II) complex Ru(CO)3Cl(glycinate) (CORM-3) on Escherichia coli By resorting to 1H nuclear magnetic resonance, mass spectrometry, and enzymatic activities, we show that CORM-3-treated E. coli accumulates larger amounts of glycolytic intermediates, independently of the oxygen growth conditions. The work provides several evidences that CORM-3 inhibits glutamate synthesis and the iron-sulfur enzymes of the tricarboxylic acid (TCA) cycle and that the glycolysis pathway is triggered in order to establish an energy and redox homeostasis balance. Accordingly, supplementation of the growth medium with fumarate, α-ketoglutarate, glutamate, and amino acids cancels the toxicity of CORM-3. Importantly, inhibition of the iron-sulfur enzymes glutamate synthase, aconitase, and fumarase is only observed for compounds that liberate carbon monoxide. Altogether, this work reveals that the antimicrobial action of CORM-3 results from intracellular glutamate deficiency and inhibition of nitrogen and TCA cycles.


Asunto(s)
Antibacterianos/farmacología , Monóxido de Carbono/farmacología , Ciclo del Ácido Cítrico/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica , Nitrógeno/metabolismo , Compuestos Organometálicos/farmacología , Aconitato Hidratasa/antagonistas & inhibidores , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Antibacterianos/química , Monóxido de Carbono/química , Ciclo del Ácido Cítrico/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fumarato Hidratasa/antagonistas & inhibidores , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Fumaratos/metabolismo , Glutamato Sintasa/antagonistas & inhibidores , Glutamato Sintasa/genética , Glutamato Sintasa/metabolismo , Ácido Glutámico/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/genética , Ácidos Cetoglutáricos/metabolismo , Espectroscopía de Resonancia Magnética , Metabolómica/métodos , Compuestos Organometálicos/química , Oxidación-Reducción
3.
J Neuroinflammation ; 14(1): 130, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28655348

RESUMEN

BACKGROUND: The endogenously produced gaseous molecule carbon monoxide is able to promote organ protection after ischemia-reperfusion injuries (IRI). The impact of carbon monoxide releasing molecules (CORM) regarding inflammation in neuronal tissues has not been studied in detail. In this investigation, we aimed to analyze the effects of the CORM ALF-186 on neuro-inflammation and hypothesized that the soluble guanylate cyclase (sGC) is playing a decisive role. METHODS: Retinal ischemia-reperfusion injury was performed for 60 min in Sprague-Dawley rats. Thereafter, the CORM ALF-186 (10 mg/kg) in the presence or absence of the sGC inhibitor ODQ was injected via a tail vein. Retinal tissue was harvested 24 h later to analyze mRNA or protein expression of sGC-ß1 subunit, transcription factors NF-κB and CREB, the inflammatory cytokines TNF-α and IL-6, as well as the heat shock proteins (HSP) HSP-70 and HSP-90. Immunohistochemistry was performed on frozen sections of the retina. The overall neuroprotective effect of ALF-186 was assessed by counting fluorogold-pre-labeled retinal ganglion cells (RGC) 7 days after IRI. RESULTS: Ischemia-reperfusion mediated loss of vital RGC was attenuated by the administration of ALF-186 after injury. ALF-186 treatment after IRI induced sGC-ß1 leading to a decreased NF-κB and CREB phosphorylation. Consecutively, ALF-186 mitigated IRI induced TNF-α and IL-6 expression in the retina and in the rats' serum. Moreover, ALF-186 attenuated heat shock protein 70 (Hsp-70) while increasing Hsp-90. The sGC-inhibitor ODQ attenuated the anti-inflammatory effects of ALF-186 and increased retinal loss of ganglion cells. These results were confirmed by immunohistochemistry. CONCLUSION: The CORM ALF-186 protected RGC from IRI induced loss. Furthermore, ALF-186 reduced IRI mediated neuroinflammation in the retina and in the serum by activating sGC. Inhibition of sGC stopped the beneficial and protective effects of ALF-186. ALF-186 may present a promising therapeutic alternative in treating inflammation after neuronal IRI.


Asunto(s)
Antiinflamatorios/uso terapéutico , Complejos de Coordinación/uso terapéutico , Isquemia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Guanilil Ciclasa Soluble/metabolismo , Animales , Antiinflamatorios/farmacología , Monóxido de Carbono/metabolismo , Complejos de Coordinación/farmacología , Femenino , Isquemia/metabolismo , Isquemia/patología , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
4.
Inorg Chem ; 56(17): 10474-10480, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28820251

RESUMEN

Mesoporous silica Al-MCM-41 nanoparticles have been used, for the first time, as vehicles for the single and dual encapsulation of the cationic CO-releasing molecule (CORM) [Mn(1,4,7-triazacyclononane)(CO)3]+ (ALF472+) and the well-known antineoplastic drug, cis-[PtCl2(NH3)2] (cisplatin). Thus, two new hybrid materials, namely, ALF472@Al-MCM-41 and ALF472-cisplatin@Al-MCM-41, have been isolated and fully characterized. The results reveal that the presence of CORM molecules enhances cisplatin loading 3-fold, yielding a cargo of 0.45 mmol g-1 of ALF472+ and 0.12 mmol g-1 of the platinum complex for ALF472-cisplatin@Al-MCM-41. It is worth noting that ALF472@Al-MCM-41 shows a good dispersion in phosphate buffered saline solution, while the dual hybrid material slightly aggregates in this simulated physiological medium (hydrodynamic size: 112 ± 23 and 336 ± 50 nm, respectively). In addition, both hybrid materials (ALF472@Al-MCM-41 and ALF472-cisplatin@Al-MCM-41) behave as photoactive CO-releasing materials, delivering 0.25 and 0.11 equiv of CO, respectively, after 24 h and exhibiting a more controlled CO delivery than that of the free CORM. Finally, metal leaching studies have confirmed the good retention capacity of Al-MCM-41 toward the potentially toxic manganese fragments (86% of retention after 72 h) as well as the low release of cisplatin (ca. 7% after 72 h).


Asunto(s)
Monóxido de Carbono/química , Cisplatino/química , Complejos de Coordinación/química , Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silicio/química , Aluminio/química , Cinética , Ligandos , Compuestos Organometálicos
5.
Inorg Chem ; 55(13): 6525-31, 2016 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-27291890

RESUMEN

The encapsulation of the photoactive, nontoxic, water-soluble, and air-stable cationic CORM [Mn(tacn)(CO)3]Br (tacn = 1,4,7-triazacyclononane) in different inorganic porous matrixes, namely, the metalorganic framework bio-MOF-1, (NH2(CH3)2)2[Zn8(adeninate)4(BPDC)6]·8DMF·11H2O (BPDC = 4,4'-biphenyldicarboxylate), and the functionalized mesoporous silicas MCM-41-SO3H and SBA-15-SO3H, is achieved by a cation exchange strategy. The CO release from these loaded materials, under simulated physiological conditions, is triggered by visible light. The results show that the silica matrixes, which are unaltered under physiological conditions, slow the kinetics of CO release, allowing a more controlled CO supply. In contrast, bio-MOF-1 instability leads to the complete leaching of the CORM. Nevertheless, the degradation of the MOF matrix gives rise to an enhanced CO release rate, which is related to the presence of free adenine in the solution.

6.
Chemistry ; 21(42): 14708-12, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26316066

RESUMEN

We have designed and synthesised a [Ru(CO)3 Cl2 (NAC)] pro-drug that features an N-acetyl cysteine (NAC) ligand. This NAC carbon monoxide releasing molecule (CORM) conjugate is able to simultaneously release biologically active CO and to ablate the concurrent formation of reactive oxygen species (ROS). Complexes of the general formulae [Ru(CO)3 (L)3 ](2+) , including [Ru(CO)3 Cl(glycinate)] (CORM-3), have been shown to produce ROS through a water-gas shift reaction, which contributes significantly, for example, to their antibacterial activity. In contrast, NAC-CORM conjugates do not produce ROS or possess antibacterial activity. In addition, we demonstrate the synergistic effect of CO and NAC both for the inhibition of nitric oxide (formation) and in the expression of tumour-necrosis factor (TNF)-α. This work highlights the advantages of combining a CO-releasing scaffold with the anti-oxidant and anti-inflammatory drug NAC in a unique pro-drug.

7.
Angew Chem Int Ed Engl ; 54(4): 1172-5, 2015 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-25477186

RESUMEN

We demonstrate that Ru(II)(CO)2-protein complexes, formed by the reaction of the hydrolytic decomposition products of [fac-RuCl(κ(2)-H2NCH2CO2)(CO)3] (CORM-3) with histidine residues exposed on the surface of proteins, spontaneously release CO in aqueous solution, cells, and mice. CO release was detected by mass spectrometry (MS) and confocal microscopy using a CO-responsive turn-on fluorescent probe. These findings support our hypothesis that plasma proteins act as CO carriers after in vivo administration of CORM-3. CO released from a synthetic bovine serum albumin (BSA)-Ru(II)(CO)2 complex leads to downregulation of the cytokines interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α in cancer cells. Finally, administration of BSA-Ru(II)(CO)2 in mice bearing a colon carcinoma tumor results in enhanced CO accumulation at the tumor. Our data suggest the use of Ru(II)(CO)2-protein complexes as viable alternatives for the safe and spatially controlled delivery of therapeutic CO in vivo.


Asunto(s)
Compuestos Organometálicos/química , Albúmina Sérica Bovina/química , Animales , Monóxido de Carbono/análisis , Monóxido de Carbono/metabolismo , Bovinos , Línea Celular Tumoral , Femenino , Células HeLa , Histidina/química , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Profármacos/química , Profármacos/metabolismo , Profármacos/uso terapéutico , Albúmina Sérica Bovina/metabolismo , Distribución Tisular , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Dalton Trans ; 53(26): 11009-11020, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38874948

RESUMEN

The toxicity profile of fac-[Re(CO)3(N-N)L]+ complexes against microbial and tumoral cells has been extensively studied, primarily focusing on modifications to the bidentate diimine (N-N) ligand. However, less attention has been paid to modifications of the axial ligand L, which is perpendicular to the Re-N-N plane. This study reveals that the high toxicity of the fac-[Re(CO)3(bpy)(Ctz)]+ complex may be attributed to the structural effect of the trityl (CPh3) group present in clotrimazole, as removal of phenyl rings causes a significant decrease in the activity against Staphylococcus aureus (S. aureus). Moreover, substitution of the 1-tritylimidazole ligand by the structurally related ligands PPh3 and PCy3 maintains similarly high activity levels. These findings contribute to understanding the interactions of toxic complexes with bacterial membranes, suggesting that the ligand structures play a crucial role in inhibiting cell wall synthesis processes, potentially including Lipid II synthesis. Compounds with Ph3E (E = C-imidazole; P) groups also showed to be 10 times more toxic than cisplatin against three mammalian cell lines (IC50: 2-4 µM). In contrast, the analogue 1-benzylimidazole and 1-tert-butylimidazole derivatives were as toxic as cisplatin. We observed that the decomposition of the [Re(I)(CO)3] fragment inside mammalian cell lines liberates CO, which is expected to exert biological effects. Therefore, compounds of this family possessing the structural motif Ph3E seem to combine high antimicrobial and antitumoral activities, the latter being much higher than that of cisplatin.


Asunto(s)
Antineoplásicos , Monóxido de Carbono , Complejos de Coordinación , Pruebas de Sensibilidad Microbiana , Renio , Staphylococcus aureus , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Staphylococcus aureus/efectos de los fármacos , Monóxido de Carbono/química , Monóxido de Carbono/farmacología , Renio/química , Renio/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Línea Celular Tumoral , Estructura Molecular , Ligandos , Ensayos de Selección de Medicamentos Antitumorales , Supervivencia Celular/efectos de los fármacos , Relación Estructura-Actividad , Proliferación Celular/efectos de los fármacos
9.
Chem Soc Rev ; 41(9): 3571-83, 2012 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-22349541

RESUMEN

The use of Carbon Monoxide (CO) as a therapeutic agent has already been tested in human clinical trials. Pre-clinically, CO gas administration proved beneficial in animal models of various human diseases. However, the use of gaseous CO faces serious obstacles not the least being its well-known toxicity. To fully realise the promise of CO as a therapeutic agent, it is key to find novel avenues for CO delivery to diseased tissues in need of treatment, without concomitant formation of elevated, toxic blood levels of carboxyhemoglobin (COHb). CO-releasing molecules (CO-RMs) have the potential to constitute safe treatments if CO release in vivo can be controlled in a spatial and temporal manner. It has already been demonstrated in animals that CO-RMs can release CO and mimic the therapeutic effects of gaseous CO. While demonstrating the principle of treatment with CO-RMs, these first generation compounds are not suitable for human use. This tutorial review summarises the biological and chemical behaviour of CO, the current status of CO-RM development, and derives principles for the creation of the next generation of CO-RMs for clinical applications in humans.


Asunto(s)
Monóxido de Carbono/química , Monóxido de Carbono/farmacología , Monóxido de Carbono/uso terapéutico , Carboxihemoglobina/química , Carboxihemoglobina/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Humanos , Hígado/efectos de los fármacos , Compuestos Organometálicos/química , Teoría Cuántica
10.
Antioxidants (Basel) ; 12(6)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37371928

RESUMEN

Carbon monoxide (CO) is a cytoprotective endogenous gas that is ubiquitously produced by the stress response enzyme heme-oxygenase. Being a gas, CO rapidly diffuses through tissues and binds to hemoglobin (Hb) increasing carboxyhemoglobin (COHb) levels. COHb can be formed in erythrocytes or in plasma from cell-free Hb. Herein, it is discussed as to whether endogenous COHb is an innocuous and inevitable metabolic waste product or not, and it is hypothesized that COHb has a biological role. In the present review, literature data are presented to support this hypothesis based on two main premises: (i) there is no direct correlation between COHb levels and CO toxicity, and (ii) COHb seems to have a direct cytoprotective and antioxidant role in erythrocytes and in hemorrhagic models in vivo. Moreover, CO is also an antioxidant by generating COHb, which protects against the pro-oxidant damaging effects of cell-free Hb. Up to now, COHb has been considered as a sink for both exogenous and endogenous CO generated during CO intoxication or heme metabolism, respectively. Hallmarking COHb as an important molecule with a biological (and eventually beneficial) role is a turning point in CO biology research, namely in CO intoxication and CO cytoprotection.

11.
J Biol Chem ; 286(30): 26708-17, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21646348

RESUMEN

CO-releasing molecules (CO-RMs) were previously shown by us to be more potent bactericides than CO gas. This suggests a mechanism of action for CO-RM, which either potentiates the activity of CO or uses another CO-RM-specific effect. We have also reported that CORM-2 induces the expression of genes related to oxidative stress. In the present study we intend to establish whether the generation of reactive oxygen species by CO-RMs may indeed result in the inhibition of bacterial cellular function. We now report that two CO-RMs (CORM-2 and ALF062) stimulate the production of ROS in Escherichia coli, an effect that is abolished by addition of antioxidants. Furthermore, deletion of genes encoding E. coli systems involved in reactive oxygen species scavenging, namely catalases and superoxide dismutases, potentiates the lethality of CORM-2 due to an increase of intracellular ROS content. CORM-2 also induces the expression of the E. coli DNA repair/SOS system recA, and its inactivation enhances toxicity of CORM-2. Moreover, fluorescence microscopy images reveal that CORM-2 causes DNA lesions to bacterial cells. We also demonstrate that cells treated with CORM-2 contain higher levels of free iron arising from destruction of iron-sulfur proteins. Importantly, we show that CO-RMs generate hydroxyl radicals in a cell-free solution, a process that is abolished by scavenging CO. Altogether, we provide a novel insight into the molecular basis of CO-RMs action by showing that their bactericidal properties are linked to cell damage inflicted by the oxidative stress that they are able to generate.


Asunto(s)
Antibacterianos/farmacocinética , Monóxido de Carbono/metabolismo , Escherichia coli K12/metabolismo , Compuestos Organometálicos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Catalasa/genética , Catalasa/metabolismo , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Estrés Oxidativo/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
12.
Antimicrob Agents Chemother ; 56(3): 1281-90, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155828

RESUMEN

Severe forms of malaria infection, such as cerebral malaria (CM) and acute lung injury (ALI), are mainly caused by the apicomplexan parasite Plasmodium falciparum. Primary therapy with quinine or artemisinin derivatives is generally effective in controlling P. falciparum parasitemia, but mortality from CM and other forms of severe malaria remains unacceptably high. Herein, we report the design and synthesis of a novel carbon monoxide-releasing molecule (CO-RM; ALF492) that fully protects mice against experimental CM (ECM) and ALI. ALF492 enables controlled CO delivery in vivo without affecting oxygen transport by hemoglobin, the major limitation in CO inhalation therapy. The protective effect is CO dependent and induces the expression of heme oxygenase-1, which contributes to the observed protection. Importantly, when used in combination with the antimalarial drug artesunate, ALF492 is an effective adjunctive and adjuvant treatment for ECM, conferring protection after the onset of severe disease. This study paves the way for the potential use of CO-RMs, such as ALF492, as adjunctive/adjuvant treatment in severe forms of malaria infection.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antimaláricos/síntesis química , Monóxido de Carbono/metabolismo , Malaria Cerebral/tratamiento farmacológico , Compuestos Organometálicos/síntesis química , Plasmodium berghei/efectos de los fármacos , Tiogalactósidos/síntesis química , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/microbiología , Animales , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Carboxihemoglobina/metabolismo , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Malaria Cerebral/metabolismo , Malaria Cerebral/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Compuestos Organometálicos/farmacología , Compuestos Organometálicos/uso terapéutico , Plasmodium berghei/fisiología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Índice de Severidad de la Enfermedad , Tiogalactósidos/farmacología , Tiogalactósidos/uso terapéutico
13.
Arterioscler Thromb Vasc Biol ; 31(11): 2570-6, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21836072

RESUMEN

OBJECTIVE: Carbon monoxide (CO) is a weak soluble guanylyl cyclase stimulator, leading to transient increases in cGMP and vasodilation. The aim of the present work was to measure the effect of CO-releasing molecules (CORMs) on the cGMP/nitric oxide (NO) pathway and to evaluate how selected CORMs affect NO-induced vasorelaxation. METHODS AND RESULTS: Incubation of smooth muscle cells with some but not all of the CORMs caused a minor increase in cGMP levels. Concentration-response curves were bell-shaped, with higher CORMs concentrations producing lower increases in cGMP levels. Although exposure of cells to CORM-2 enhanced cGMP formation, we observed that the compound inhibited NO-stimulated cGMP accumulation in cells and NO-stimulated soluble guanylyl cyclase activity that could be reversed by superoxide anion scavengers. Reactive oxygen species generation from CORMs was confirmed using luminol-induced chemiluminescence and electron spin resonance. Furthermore, we observed that NO is scavenged by CORM-2. When used alone CORM-2 relaxed vessels through a cGMP-mediated pathway but attenuated NO donor-stimulated vasorelaxation. CONCLUSION: We conclude that the CORMs examined have context-dependent effects on vessel tone, as they can directly dilate blood vessels, but also block NO-induced vasorelaxation.


Asunto(s)
Aorta/efectos de los fármacos , Monóxido de Carbono/farmacología , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/farmacología , Vasodilatación/efectos de los fármacos , Animales , Aorta/citología , Aorta/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Guanilato Ciclasa/metabolismo , Masculino , Modelos Animales , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vasodilatación/fisiología
14.
Mol Neurobiol ; 59(2): 916-931, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34797521

RESUMEN

Microglia are the immune competent cell of the central nervous system (CNS), promoting brain homeostasis and regulating inflammatory response against infection and injury. Chronic or exacerbated neuroinflammation is a cause of damage in several brain pathologies. Endogenous carbon monoxide (CO), produced from the degradation of heme, is described as anti-apoptotic and anti-inflammatory in several contexts, including in the CNS. Neuroglobin (Ngb) is a haemoglobin-homologous protein, which upregulation triggers antioxidant defence and prevents neuronal apoptosis. Thus, we hypothesised a crosstalk between CO and Ngb, in particular, that the anti-neuroinflammatory role of CO in microglia depends on Ngb. A novel CO-releasing molecule (ALF826) based on molybdenum was used for delivering CO in microglial culture.BV-2 mouse microglial cell line was challenged with lipopolysaccharide (LPS) for triggering inflammation, and after 6 h ALF826 was added. CO exposure limited inflammation by decreasing inducible nitric oxide synthase (iNOS) expression and the production of nitric oxide (NO) and tumour necrosis factor-α (TNF-α), and by increasing interleukine-10 (IL-10) release. CO-induced Ngb upregulation correlated in time with CO's anti-inflammatory effect. Moreover, knocking down Ngb reversed the anti-inflammatory effect of CO, suggesting that dependents on Ngb expression. CO-induced Ngb upregulation was independent on ROS signalling, but partially dependent on the transcriptional factor SP1. Finally, microglial cell metabolism is also involved in the inflammatory response. In fact, LPS treatment decreased oxygen consumption in microglia, indicating a switch to glycolysis, which is associated with a proinflammatory. While CO treatment increased oxygen consumption, reverting LPS effect and indicating a metabolic shift into a more oxidative metabolism. Moreover, in the absence of Ngb, this phenotype was no longer observed, indicating Ngb is needed for CO's modulation of microglial metabolism. Finally, the metabolic shift induced by CO did not depend on alteration of mitochondrial population. In conclusion, neuroglobin emerges for the first time as a key player for CO signalling against exacerbated inflammation in microglia.


Asunto(s)
Monóxido de Carbono , Microglía , Animales , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacología , Inflamación/patología , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Neuroglobina/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo
15.
Mol Neurobiol ; 59(2): 872-889, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34796462

RESUMEN

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.


Asunto(s)
Microglía , Fármacos Neuroprotectores , Animales , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacología , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas
16.
ACS Bio Med Chem Au ; 2(4): 419-436, 2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35996473

RESUMEN

Several metal-based carbon monoxide-releasing molecules (CORMs) are active CO donors with established antibacterial activity. Among them, CORM conjugates with azole antibiotics of type [Mn(CO)3(2,2'-bipyridyl)(azole)]+ display important synergies against several microbes. We carried out a structure-activity relationship study based upon the lead structure of [Mn(CO)3(Bpy)(Ctz)]+ by producing clotrimazole (Ctz) conjugates with varying metal and ligands. We concluded that the nature of the bidentate ligand strongly influences the bactericidal activity, with the substitution of bipyridyl by small bicyclic ligands leading to highly active clotrimazole conjugates. On the contrary, the metal did not influence the activity. We found that conjugate [Re(CO)3(Bpy)(Ctz)]+ is more than the sum of its parts: while precursor [Re(CO)3(Bpy)Br] has no antibacterial activity and clotrimazole shows only moderate minimal inhibitory concentrations, the potency of [Re(CO)3(Bpy)(Ctz)]+ is one order of magnitude higher than that of clotrimazole, and the spectrum of bacterial target species includes Gram-positive and Gram-negative bacteria. The addition of [Re(CO)3(Bpy)(Ctz)]+ to Staphylococcus aureus causes a general impact on the membrane topology, has inhibitory effects on peptidoglycan biosynthesis, and affects energy functions. The mechanism of action of this kind of CORM conjugates involves a sequence of events initiated by membrane insertion, followed by membrane disorganization, inhibition of peptidoglycan synthesis, CO release, and break down of the membrane potential. These results suggest that conjugation of CORMs to known antibiotics may produce useful structures with synergistic effects that increase the conjugate's activity relative to that of the antibiotic alone.

17.
J Am Chem Soc ; 133(5): 1192-5, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21204537

RESUMEN

CORM-3, [fac-Ru(CO)(3)Cl(κ(2)-H(2)NCH(2)CO(2))], is a well-known carbon monoxide releasing molecule (CORM) capable of delivering CO in vivo. Herein we show for the first time that the interactions of CORM-3 with proteins result in the loss of a chloride ion, glycinate, and one CO ligand. The rapid formation of stable adducts between the protein and the remaining cis-Ru(II)(CO)(2) fragments was confirmed by Inductively Coupled Plasma-Atomic Emission Spectroscopy (ICP-AES), Liquid-Chromatography Mass Spectrometry (LC-MS), Infrared Spectroscopy (IR), and X-ray crystallography. Three Ru coordination sites are observed in the structure of hen egg white lysozyme crystals soaked with CORM-3. The site with highest Ru occupancy (80%) shows a fac-[(His15)Ru(CO)(2)(H(2)O)(3)] structure.


Asunto(s)
Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Muramidasa/química , Muramidasa/metabolismo , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Animales , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Conformación Proteica
18.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 2): m288-9, 2011 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-21522931

RESUMEN

The title compound, [Mo(2)(C(5)H(10)NS(2))(2)O(4)]·C(4)H(8)O, can be readily prepared in tetra-hydro-furan (THF) by an oxidation reaction between the Mo(IV) precursor [MoO(S(2)CNEt(2))(2)] with [ReMeO(3)]. The compound is an axially symmetric Mo(V) dimer (2 symmetry), in which the metal atoms exhibit a distorted square-pyramidal coordination environment. A THF mol-ecule was found to be equally disordered over two symmetry-related sites (around a twofold rotation axis), trans-coordinated to the apical oxido group and weakly inter-acting with the Mo(V) atoms [Mo-O = 2.6213 (19) Å]. In the crystal, some weak C-H⋯O inter-actions occur between the terminal oxido and neighbouring -CH(3) groups of an adjacent [Mo(µ-O)O(S(2)CNEt(2))](2) unit.

19.
ACS Biomater Sci Eng ; 6(2): 1090-1101, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33464857

RESUMEN

CO-releasing molecules (CORMs) have been widely studied for their anti-inflammatory, antiapoptotic, and antiproliferative effects. CORM-3 is a water-soluble Ru-based metal carbonyl complex, which metallates serum proteins and readily releases CO in biological media. In this work, we evaluated the anti-inflammatory and wound-healing effects of gold nanoparticles-CORM-3 conjugates, AuNPs@PEG@BSA·Ru(CO)x, exploring its use as an efficient CO carrier. Our results suggest that the nanoformulation was capable of inducing a more pronounced cell effect, at the anti-inflammatory level and a faster tissue repair, probably derived from a rapid cell uptake of the nanoformulation that results in the increase of CO inside the cell.


Asunto(s)
Complejos de Coordinación , Nanopartículas del Metal , Compuestos Organometálicos , Monóxido de Carbono , Oro
20.
J Org Chem ; 74(18): 6960-4, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19685891

RESUMEN

The reduction of aromatic nitro compounds to the corresponding amines with silanes catalyzed by high valent oxo-rhenium complexes is reported. The catalytic systems PhMe(2)SiH/ReIO(2)(PPh(3))(2) (5 mol %) and PhMe(2)SiH/ReOCl(3)(PPh(3))(2) (5 mol %) reduced efficiently a series of aromatic nitro compounds in the presence of a wide range of functional groups such as ester, halo, amide, sulfone, lactone, and benzyl. This methodology also allowed the regioselective reduction of dinitrobenzenes to the corresponding nitroanilines and the reduction of an aromatic nitro group in presence of an aliphatic nitro group.


Asunto(s)
Hidrocarburos Aromáticos/química , Nitrocompuestos/química , Compuestos Organometálicos/química , Renio/química , Silanos/química , Siloxanos/química , Compuestos de Anilina/química , Catálisis , Dinitrobencenos/química , Modelos Químicos , Oxidación-Reducción , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA