Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arthritis Rheumatol ; 76(2): 279-284, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37695218

RESUMEN

OBJECTIVE: Calcium pyrophosphate (CPP) crystal deposition in the joints is associated with a heterogeneous set of debilitating syndromes characterized by inflammation and pain, for which no effective therapies are currently available. Because we found that the mitochondrial enzyme monoamine oxidase B (MAO-B) plays a fundamental role in promoting inflammatory pathways, this study aims at assessing the efficacy of two clinical-grade inhibitors (iMAO-Bs) in preclinical models of this disease to pave the way for a novel treatment. METHODS: We tested our hypothesis in two murine models of CPP-induced arthritis, by measuring cytokine and chemokine levels, along with immune cell recruitment. iMAO-Bs (rasagiline and safinamide) were administered either before or after crystal injection. To elucidate the molecular mechanism, we challenged in vitro primed macrophages with CPP crystals and assessed the impact of iMAO-Bs in dampening proinflammatory cytokines and in preserving mitochondrial function. RESULTS: Both in preventive and therapeutic in vivo protocols, iMAO-Bs blunted the release of proinflammatory cytokines (interleukin [IL]-6 and IL1-ß) and chemokines (CXCL10, CXCL1, CCL2 and CCL5) (n > 6 mice/group). Importantly, they also significantly reduced ankle swelling (50.3% vs 17.1%; P < 0.001 and 23.1%; P = 0.005 for rasagiline and safinamide, respectively). Mechanistically, iMAO-Bs dampened the burst of reactive oxygen species and the mitochondrial dysfunction triggered by CPP crystals in isolated macrophages. Moreover, iMAO-Bs blunted cytokine secretion and NLRP3 inflammasome activation through inhibition of the NF-κB and STAT3 pathways. CONCLUSION: iMAO-Bs dampen inflammation in murine models of crystal-induced arthropathy, thereby uncovering MAO-B as a promising target to treat these diseases.


Asunto(s)
Alanina/análogos & derivados , Artritis , Bencilaminas , Pirofosfato de Calcio , Indanos , Ratones , Animales , Monoaminooxidasa/metabolismo , Citocinas , Inflamación/metabolismo , Artritis/metabolismo , Quimiocinas/metabolismo , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Mitocondrias/metabolismo , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
2.
Cell Death Differ ; 30(3): 742-752, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36307526

RESUMEN

Macrophages are essential players for the host response against pathogens, regulation of inflammation and tissue regeneration. The wide range of macrophage functions rely on their heterogeneity and plasticity that enable a dynamic adaptation of their responses according to the surrounding environmental cues. Recent studies suggest that metabolism provides synergistic support for macrophage activation and elicitation of desirable immune responses; however, the metabolic pathways orchestrating macrophage activation are still under scrutiny. Optic atrophy 1 (OPA1) is a mitochondria-shaping protein controlling mitochondrial fusion, cristae biogenesis and respiration; clear evidence shows that the lack or dysfunctional activity of this protein triggers the accumulation of metabolic intermediates of the TCA cycle. In this study, we show that OPA1 has a crucial role in macrophage activation. Selective Opa1 deletion in myeloid cells impairs M1-macrophage commitment. Mechanistically, Opa1 deletion leads to TCA cycle metabolite accumulation and defective NF-κB signaling activation. In an in vivo model of muscle regeneration upon injury, Opa1 knockout macrophages persist within the damaged tissue, leading to excess collagen deposition and impairment in muscle regeneration. Collectively, our data indicate that OPA1 is a key metabolic driver of macrophage functions.


Asunto(s)
Mitocondrias , Membranas Mitocondriales , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Transducción de Señal , Macrófagos/metabolismo
3.
Front Bioeng Biotechnol ; 10: 941623, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35898644

RESUMEN

Skeletal muscle is a fundamental tissue of the human body with great plasticity and adaptation to diseases and injuries. Recreating this tissue in vitro helps not only to deepen its functionality, but also to simulate pathophysiological processes. In this review we discuss the generation of human skeletal muscle three-dimensional (3D) models obtained through tissue engineering approaches. First, we present an overview of the most severe myopathies and the two key players involved: the variety of cells composing skeletal muscle tissue and the different components of its extracellular matrix. Then, we discuss the peculiar characteristics among diverse in vitro models with a specific focus on cell sources, scaffold composition and formulations, and fabrication techniques. To conclude, we highlight the efficacy of 3D models in mimicking patient-specific myopathies, deepening muscle disease mechanisms or investigating possible therapeutic effects.

4.
Cancers (Basel) ; 13(21)2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34771641

RESUMEN

Macrophages are immune cells that are important for the development of the defensive front line of the innate immune system. Following signal recognition, macrophages undergo activation toward specific functional states, consisting not only in the acquisition of specific features but also of peculiar metabolic programs associated with each function. For these reasons, macrophages are often isolated from mice to perform cellular assays to study the mechanisms mediating immune cell activation. This requires expensive and time-consuming breeding and housing of mice strains. To overcome this issue, we analyzed an in-house J2-generated immortalized macrophage cell line from BMDMs, both from a functional and metabolic point of view. By assaying the intracellular and extracellular metabolism coupled with the phenotypic features of immortalized versus primary BMDMs, we concluded that classically and alternatively immortalized macrophages display similar phenotypical, metabolic and functional features compared to primary cells polarized in the same way. Our study validates the use of this immortalized cell line as a suitable model with which to evaluate in vitro how perturbations can influence the phenotypical and functional features of murine macrophages.

5.
Front Immunol ; 12: 734229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659222

RESUMEN

Reactive oxygen species (ROS) are fundamental for macrophages to eliminate invasive microorganisms. However, as observed in nonphagocytic cells, ROS play essential roles in processes that are different from pathogen killing, as signal transduction, differentiation, and gene expression. The different outcomes of these events are likely to depend on the specific subcellular site of ROS formation, as well as the duration and extent of ROS production. While excessive accumulation of ROS has long been appreciated for its detrimental effects, there is now a deeper understanding of their roles as signaling molecules. This could explain the failure of the "all or none" pharmacologic approach with global antioxidants to treat several diseases. NADPH oxidase is the first source of ROS that has been identified in macrophages. However, growing evidence highlights mitochondria as a crucial site of ROS formation in these cells, mainly due to electron leakage of the respiratory chain or to enzymes, such as monoamine oxidases. Their role in redox signaling, together with their exact site of formation is only partially elucidated. Hence, it is essential to identify the specific intracellular sources of ROS and how they influence cellular processes in both physiological and pathological conditions to develop therapies targeting oxidative signaling networks. In this review, we will focus on the different sites of ROS formation in macrophages and how they impact on metabolic processes and inflammatory signaling, highlighting the role of mitochondrial as compared to non-mitochondrial ROS sources.


Asunto(s)
Macrófagos/enzimología , Mitocondrias/enzimología , Oxidorreductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Animales , Metabolismo Energético , Humanos , Mediadores de Inflamación/metabolismo , Oxidación-Reducción
6.
EMBO Rep ; 22(9): e51981, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34260142

RESUMEN

Glutaminolysis is known to correlate with ovarian cancer aggressiveness and invasion. However, how this affects the tumor microenvironment is elusive. Here, we show that ovarian cancer cells become addicted to extracellular glutamine when silenced for glutamine synthetase (GS), similar to naturally occurring GS-low, glutaminolysis-high ovarian cancer cells. Glutamine addiction elicits a crosstalk mechanism whereby cancer cells release N-acetylaspartate (NAA) which, through the inhibition of the NMDA receptor, and synergistically with IL-10, enforces GS expression in macrophages. In turn, GS-high macrophages acquire M2-like, tumorigenic features. Supporting this in␣vitro model, in silico data and the analysis of ascitic fluid isolated from ovarian cancer patients prove that an M2-like macrophage phenotype, IL-10 release, and NAA levels positively correlate with disease stage. Our study uncovers the unprecedented role of glutamine metabolism in modulating macrophage polarization in highly invasive ovarian cancer and highlights the anti-inflammatory, protumoral function of NAA.


Asunto(s)
Ácido Aspártico , Neoplasias Ováricas , Ácido Aspártico/análogos & derivados , Línea Celular Tumoral , Femenino , Humanos , Macrófagos , Neoplasias Ováricas/genética , Microambiente Tumoral
8.
Pharmacol Ther ; 210: 107521, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32151665

RESUMEN

From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.


Asunto(s)
Metabolismo Energético , Macrófagos/metabolismo , Metaboloma , Animales , Antiinflamatorios/uso terapéutico , Antineoplásicos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Linaje de la Célula , Metabolismo Energético/efectos de los fármacos , Humanos , Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Metaboloma/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
9.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801292

RESUMEN

: Duchenne muscular dystrophy (DMD) is one of the most severe forms of inherited muscular dystrophies. The disease is caused by the lack of dystrophin, a structurally essential protein; hence, a definitive cure would necessarily have to pass through some form of gene and/or cell therapy. Cell- and genetic-based therapeutics for DMD have been explored since the 1990s and recently, two of the latter have been approved for clinical use, but their efficacy is still very low. In parallel, there have been great ongoing efforts aimed at targeting the downstream pathogenic effects of dystrophin deficiency using classical pharmacological approaches, with synthetic or biological molecules. However, as it is always the case with rare diseases, R&D costs for new drugs can represent a major hurdle for researchers and patients alike. This problem can be greatly alleviated by experimenting the use of molecules that had originally been developed for different conditions, a process known as drug repurposing or drug repositioning. In this review, we will describe the state of the art of such an approach for DMD, both in the context of clinical trials and pre-clinical studies.


Asunto(s)
Reposicionamiento de Medicamentos/métodos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Fármacos Neuromusculares/uso terapéutico , Prednisona/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Distrofina/deficiencia , Distrofina/genética , Gentamicinas/uso terapéutico , Humanos , Metformina/uso terapéutico , Ratones Transgénicos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Pregnenodionas/uso terapéutico , Simvastatina/uso terapéutico , Tadalafilo/uso terapéutico , Tamoxifeno/uso terapéutico
10.
Oxid Med Cell Longev ; 2019: 3904905, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379988

RESUMEN

Coenzyme Q (CoQ), a redox-active lipid, is comprised of a quinone group and a polyisoprenoid tail. It is an electron carrier in the mitochondrial respiratory chain, a cofactor of other mitochondrial dehydrogenases, and an essential antioxidant. CoQ requires a large set of enzymes for its biosynthesis; mutations in genes encoding these proteins cause primary CoQ deficiency, a clinically and genetically heterogeneous group of diseases. Patients with CoQ deficiency often respond to oral CoQ10 supplementation. Treatment is however problematic because of the low bioavailability of CoQ10 and the poor tissue delivery. In recent years, bypass therapy using analogues of the precursor of the aromatic ring of CoQ has been proposed as a promising alternative. We have previously shown using a yeast model that vanillic acid (VA) can bypass mutations of COQ6, a monooxygenase required for the hydroxylation of the C5 carbon of the ring. In this work, we have generated a human cell line lacking functional COQ6 using CRISPR/Cas9 technology. We show that these cells cannot synthesize CoQ and display severe ATP deficiency. Treatment with VA can recover CoQ biosynthesis and ATP production. Moreover, these cells display increased ROS production, which is only partially corrected by exogenous CoQ, while VA restores ROS to normal levels. Furthermore, we show that these cells accumulate 3-decaprenyl-1,4-benzoquinone, suggesting that in mammals, the decarboxylation and C1 hydroxylation reactions occur before or independently of the C5 hydroxylation. Finally, we show that COQ6 isoform c (transcript NM_182480) does not encode an active enzyme. VA can be produced in the liver by the oxidation of vanillin, a nontoxic compound commonly used as a food additive, and crosses the blood-brain barrier. These characteristics make it a promising compound for the treatment of patients with CoQ deficiency due to COQ6 mutations.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ubiquinona/análogos & derivados , Ácido Vanílico/farmacología , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas/genética , Células HEK293 , Humanos , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo , Alineación de Secuencia , Ubiquinona/biosíntesis , Ubiquinona/genética , Ubiquinona/metabolismo
11.
Front Physiol ; 9: 1087, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30154729

RESUMEN

Oxidative stress and mitochondrial dysfunction play a crucial role in the pathophysiology of muscular dystrophies. We previously reported that the mitochondrial enzyme monoamine oxidase (MAO) is a relevant source of reactive oxygen species (ROS) not only in murine models of muscular dystrophy, in which it directly contributes to contractile impairment, but also in muscle cells from collagen VI-deficient patients. Here, we now assessed the efficacy of a novel MAO-B inhibitor, safinamide, using in vivo and in vitro models of Duchenne muscular dystrophy (DMD). Specifically, we found that administration of safinamide in 3-month-old mdx mice reduced myofiber damage and oxidative stress and improved muscle functionality. In vitro studies with myogenic cultures from mdx mice and DMD patients showed that even cultured dystrophic myoblasts were more susceptible to oxidative stress than matching cells from healthy donors. Indeed, upon exposure to the MAO substrate tyramine or to hydrogen peroxide, DMD muscle cells displayed a rise in ROS levels and a consequent mitochondrial depolarization. Remarkably, both phenotypes normalized when cultures were treated with safinamide. Given that safinamide is already in clinical use for neurological disorders, our findings could pave the way toward a promising translation into clinical trials for DMD patients as a classic case of drug repurposing.

12.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 3050-3059, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29953926

RESUMEN

Monoamine oxidase (MAO), a mitochondrial enzyme that oxidizes biogenic amines generating hydrogen peroxide, is a major source of oxidative stress in cardiac injury. However, the molecular mechanisms underlying its overactivation in pathological conditions are still poorly characterized. Here, we investigated whether the enhanced MAO-dependent hydrogen peroxide production can be due to increased substrate availability using a metabolomic profiling method. We identified N1-methylhistamine -the main catabolite of histamine- as an important substrate fueling MAO in Langendorff mouse hearts, directly perfused with a buffer containing hydrogen peroxide or subjected to ischemia/reperfusion protocol. Indeed, when these hearts were pretreated with the MAO inhibitor pargyline we observed N1-methylhistamine accumulation along with reduced oxidative stress. Next, we showed that synaptic terminals are the major source of N1-methylhistamine. Indeed, in vivo sympathectomy caused a decrease of N1-methylhistamine levels, which was associated with a marked protection in post-ischemic reperfused hearts. As far as the mechanism is concerned, we demonstrate that exogenous histamine is transported into isolated cardiomyocytes and triggers a rise in the levels of reactive oxygen species (ROS). Once again, pargyline pretreatment induced intracellular accumulation of N1-methylhistamine along with decrease in ROS levels. These findings uncover a receptor-independent mechanism for histamine in cardiomyocytes. In summary, our study reveals a novel and important pathophysiological causative link between MAO activation and histamine availability during pathophysiological conditions such as oxidative stress/cardiac injury.


Asunto(s)
Ventrículos Cardíacos/patología , Histamina/metabolismo , Monoaminooxidasa/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Animales , Modelos Animales de Enfermedad , Ventrículos Cardíacos/citología , Humanos , Preparación de Corazón Aislado , Masculino , Metabolómica , Metilhistaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Daño por Reperfusión Miocárdica/etiología , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción , Estrés Oxidativo , Pargilina/farmacología , Especies Reactivas de Oxígeno/metabolismo
13.
Anal Chem ; 90(9): 5687-5695, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29595056

RESUMEN

Analyses of cellular responses to fast oxygen dynamics are challenging and require ad hoc technological solutions, especially when decoupling from liquid media composition is required. In this work, we present a microfluidic device specifically designed for culture analyses with high resolution and magnification objectives, providing full optical access to the cell culture chamber. This feature allows fluorescence-based assays, photoactivated surface chemistry, and live cell imaging under tightly controlled pO2 environments. The device has a simple design, accommodates three independent cell cultures, and can be employed by users with basic cell culture training in studies requiring fast oxygen dynamics, defined media composition, and in-line data acquisition with optical molecular probes. We apply this technology to produce an oxygen/glucose deprived (OGD) environment and analyze cell mortality in murine and human cardiac cultures. Neonatal rat ventricular cardiomyocytes show an OGD time-dependent sensitivity, resulting in a robust and reproducible 66 ± 5% death rate after 3 h of stress. Applying an equivalent stress to human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) provides direct experimental evidence for fetal-like OGD-resistant phenotype. Investigation on the nature of such phenotype exposed large glycogen deposits. We propose a culture strategy aimed at depleting these intracellular energy stores and concurrently activate positive regulation of aerobic metabolic molecular markers. The observed process, however, is not sufficient to induce an OGD-sensitive phenotype in hiPS-CMs, highlighting defective development of mature aerobic metabolism in vitro.


Asunto(s)
Glucosa/análisis , Células Madre Pluripotentes Inducidas/química , Técnicas Analíticas Microfluídicas , Imagen Óptica , Oxígeno/análisis , Animales , Células Cultivadas , Glucosa/deficiencia , Glucosa/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Nat Prod Res ; 32(12): 1395-1401, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28691859

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder bearing motor and nonmotor symptoms. The treatment today is symptomatical rather than preventive or curative and this leaves the field open for the search of both novel molecular targets and drug candidates. Interference with α-synuclein fibrillation, monoamine oxidase (MAO) inhibition, modulation of adenosine receptors and the inhibition of specific phosphodiesterase (PDE) isoforms are some of the currently pursued strategies. We synthesised and studied some semi-synthetic berberine derivatives using a set of in silico tools. We evaluated their drug-likeness and tested the compounds against a set of target proteins involved in the onset or progression of PD, with a particular attention to MAO-B. Preliminary in vitro assay on MAO-B confirmed our in silico predictions.


Asunto(s)
Antiparkinsonianos/química , Antiparkinsonianos/farmacología , Berberina/química , Inhibidores de la Monoaminooxidasa/farmacología , Berberina/farmacología , Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Humanos , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida , Monoaminooxidasa/química , Monoaminooxidasa/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico
15.
Proc Natl Acad Sci U S A ; 113(46): 13009-13014, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799519

RESUMEN

We identify a target for treating obesity and type 2 diabetes, the consumption of calories by an increase in the metabolic rate of resting skeletal muscle. The metabolic rate of skeletal muscle can be increased by shifting myosin heads from the super-relaxed state (SRX), with a low ATPase activity, to a disordered relaxed state (DRX), with a higher ATPase activity. The shift of myosin heads was detected by a change in fluorescent intensity of a probe attached to the myosin regulatory light chain in skinned skeletal fibers, allowing us to perform a high-throughput screen of 2,128 compounds. The screen identified one compound, which destabilized the super-relaxed state, piperine (the main alkaloid component of black pepper). Destabilization of the SRX by piperine was confirmed by single-nucleotide turnover measurements. The effect was only observed in fast twitch skeletal fibers and not in slow twitch fibers or cardiac tissues. Piperine increased ATPase activity of skinned relaxed fibers by 66 ± 15%. The Kd was ∼2 µM. Piperine had little effect on the mechanics of either fully active or resting muscle fibers. Previous work has shown that piperine can mitigate both obesity and type 2 diabetes in rodent models of these conditions. We propose that the increase in resting muscle metabolism contributes to these positive effects. The results described here show that up-regulation of resting muscle metabolism could treat obesity and type 2 diabetes and that piperine would provide a useful lead compound for the development of these therapies.


Asunto(s)
Alcaloides/farmacología , Metabolismo Basal/efectos de los fármacos , Benzodioxoles/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Obesidad/metabolismo , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Adenosina Trifosfatasas/metabolismo , Alcaloides/uso terapéutico , Animales , Benzodioxoles/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento , Fibras Musculares de Contracción Rápida/metabolismo , Obesidad/tratamiento farmacológico , Piperidinas/uso terapéutico , Alcamidas Poliinsaturadas/uso terapéutico , Conejos , Miosinas del Músculo Esquelético/metabolismo , Regulación hacia Arriba
16.
PLoS One ; 11(8): e0160100, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27479128

RESUMEN

In the super-relaxed state of myosin, ATPase activity is strongly inhibited by binding of the myosin heads to the core of the thick filament in a structure known as the interacting-heads motif. In the disordered relaxed state myosin heads are not bound to the core of the thick filament and have an ATPase rate that is 10 fold greater. In the interacting-heads motif the two regulatory light chains appear to bind to each other. We have made single cysteine mutants of the regulatory light chain, placed both paramagnetic and fluorescent probes on them, and exchanged them into skinned skeletal muscle fibers. Many of the labeled light chains tended to disrupt the stability of the super-relaxed state, and showed spectral changes in the transition from the disordered relaxed state to the super-relaxed state. These data support the putative interface between the two regulatory light chains identified by cryo electron microscopy and show that both the divalent cation bound to the regulatory light chain and the N-terminus of the regulatory light chain play a role in the stability of the super-relaxed state. One probe showed a shift to shorter wavelengths in the super-relaxed state such that a ratio of intensities at 440nm to that at 520nm provided a measure of the population of the super-relaxed state amenable for high throughput screens for finding potential pharmaceuticals. The results provide a proof of concept that small molecules that bind to this region can destabilize the super-relaxed state and provide a method to search for small molecules that do so leading to a potentially effective treatment for Type 2 diabetes and obesity.


Asunto(s)
Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Relajación Muscular/fisiología , Adenosina Trifosfato/metabolismo , Animales , Microscopía por Crioelectrón , Espectroscopía de Resonancia por Spin del Electrón , Colorantes Fluorescentes/química , Ratones , Ratones Endogámicos C57BL , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Estructura Cuaternaria de Proteína , Conejos , Espectrometría de Fluorescencia
17.
J Muscle Res Cell Motil ; 35(1): 23-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24619215

RESUMEN

Muscular dystrophies (MDs) are a heterogeneous group of diseases that share a common end-point represented by muscular wasting. MDs are caused by mutations in a variety of genes encoding for different molecules, including extracellular matrix, transmembrane and membrane-associated proteins, cytoplasmic enzymes and nuclear proteins. However, it is still to be elucidated how genetic mutations can affect the molecular mechanisms underlying the contractile impairment occurring in these complex pathologies. The intracellular accumulation of reactive oxygen species (ROS) is widely accepted to play a key role in contractile derangements occurring in the different forms of MDs. However, scarce information is available concerning both the most relevant sources of ROS and their major molecular targets. This review focuses on (i) the sources of ROS, with a special emphasis on monoamine oxidase, a mitochondrial enzyme, and (ii) the targets of ROS, highlighting the relevance of the oxidative modification of myofilament proteins.


Asunto(s)
Distrofias Musculares/metabolismo , Estrés Oxidativo/fisiología , Animales , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Miofibrillas/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
18.
J Am Coll Cardiol ; 57(3): 300-9, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21232667

RESUMEN

OBJECTIVES: We investigated the incidence and contribution of the oxidation/nitrosylation of tropomyosin and actin to the contractile impairment and cardiomyocyte injury occurring in human end-stage heart failure (HF) as compared with nonfailing donor hearts. BACKGROUND: Although there is growing evidence that augmented intracellular accumulation of reactive oxygen/nitrogen species may play a key role in causing contractile dysfunction, there is a dearth of data regarding their contractile protein targets in human HF. METHODS: In left ventricular (LV) biopsies from explanted failing hearts (New York Heart Association functional class IV; HF group) and nonfailing donor hearts (NF group), carbonylation of actin and tropomyosin, disulphide cross-bridge (DCB) formation, and S-nitrosylation in tropomyosin were assessed, along with plasma troponin I and LV ejection fraction (LVEF). RESULTS: The LV biopsies from the HF group had 2.14 ± 0.23-fold and 2.31 ± 0.46-fold greater levels in actin and tropomyosin carbonylation, respectively, and 1.77 ± 0.45-fold greater levels of high-molecular-weight complexes of tropomyosin due to DCB formation, compared with the NF group. Tropomyosin also underwent S-nitrosylation that was 1.3 ± 0.15-fold higher in the HF group. Notably, actin and tropomyosin carbonylation was significantly correlated with both loss of viability indicated by plasma troponin I and contractile impairment as shown by reduced LVEF. CONCLUSIONS: This study demonstrated that oxidative/nitrosylative changes of actin and tropomyosin are largely increased in human failing hearts. Because these changes are inversely correlated to LVEF, actin and tropomyosin oxidation are likely to contribute to the contractile impairment evident in end-stage HF.


Asunto(s)
Actinas/metabolismo , Insuficiencia Cardíaca/metabolismo , Estrés Oxidativo/fisiología , Tropomiosina/metabolismo , Adulto , Anciano , Proteínas Contráctiles/metabolismo , Femenino , Insuficiencia Cardíaca/patología , Humanos , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
19.
Antioxid Redox Signal ; 14(5): 881-91, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20615074

RESUMEN

Mitochondrial damage is a determining factor in causing loss of cardiomyocyte function and viability, yet a mild degree of mitochondrial dysfunction appears to underlie cardioprotection against injury caused by postischemic reperfusion. This review is focused on two major mechanisms of mitochondrial dysfunction, namely, oxidative stress and opening of the mitochondrial permeability transition pore. The formation of reactive oxygen species in mitochondria will be analyzed with regard to factors controlling mitochondrial permeability transition pore opening. Finally, these mitochondrial processes are analyzed with respect to cardioprotection afforded by ischemic pre- and postconditioning.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Mitocondrias Cardíacas/patología , Animales , Humanos , Isquemia/metabolismo , Isquemia/fisiopatología , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Especies Reactivas de Oxígeno/metabolismo
20.
Biochemistry ; 49(39): 8658-67, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20804154

RESUMEN

α-Lactalbumin (LA) forms with oleic acid (OA) a complex which has been reported to induce the selective death of tumor cells. However, the mechanism by which this complex kills a wide range of tumor cell lines is as yet largely unknown. The difficulty in rationalizing the cytotoxic effects of the LA/OA complex can be due to the fact that the molecular aspects of the interaction between the protein and the fatty acid are still poorly understood, in particular regarding the oligomeric state of the protein and the actual molar ratio of OA over protein in the complex. Here, the effect of LA addition to an OA aqueous solution has been examined by dynamic light scattering measurements and transmission electron microscopy. Upon protein addition, the aggregation state of the rather insoluble OA is dramatically changed, and more water-soluble and smaller aggregates of the fatty acid are formed. A mixture of LA and an excess of OA forms a high molecular weight complex that can be isolated by size-exclusion chromatography and that displays cellular toxicity toward Jurkat cells. On the basis of gel filtration data, cross-linking experiments with glutaraldehyde, and OA titration, we evaluated that the isolated LA/OA complex is given by 4-5 protein molecules that bind 68-85 OA molecules. The protein in the complex adopts a molten globule-like conformation, and it interacts with the fatty acid mostly through its α-helical domain, as indicated by circular dichroism measurements and limited proteolysis experiments. Overall, we interpret our and previous data as indicating that the cellular toxicity of a LA/OA complex is due to the effect of a protein moiety in significantly enhancing the water solubility of the cytotoxic OA and, therefore, that the protein/OA complex can serve mainly as a carrier of the toxic fatty acid in a physiological milieu.


Asunto(s)
Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Lactalbúmina/farmacología , Ácido Oléico/metabolismo , Ácido Oléico/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Bovinos , Reactivos de Enlaces Cruzados , Humanos , Células Jurkat , Lactalbúmina/química , Lactalbúmina/metabolismo , Leucemia de Células T/tratamiento farmacológico , Conformación Molecular , Ácido Oléico/química , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...