Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Biochem Soc Trans ; 52(3): 1265-1274, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38661212

RESUMEN

The three isoforms of glycogen phosphorylase - PYGM, PYGB, and PYGL - are expressed in glial cells. Unlike PYGB and PYGL, PYGM is the only isoform regulated by Rac1. This specific regulation may confer a differential functional role compared with the other glycogen phosphorylases-PYGB and PYGL. The involvement of muscle glycogen phosphorylase in glial cells and its association with post-translational modifications (PTMs) of proteins through O-glycosylation is indeed a fascinating and emerging area of research. The dual role it plays in metabolic processes and the regulation of PTMs within the brain presents intriguing implications for various neurological conditions. Disruptions in the O-GlcNAcylation cycle and neurodegenerative diseases like Alzheimer's disease (AD) is particularly noteworthy. The alterations in O-GlcNAcylation levels of specific proteins, such as APP, c-Fos, and tau protein, highlight the intricate relationship between PTMs and AD. Understanding these processes and the regulatory function of muscle glycogen phosphorylase sheds light on its impact on protein function, signaling pathways, cellular homeostasis, neurological health, and potential interventions for brain-related conditions.


Asunto(s)
Neuroglía , Humanos , Neuroglía/metabolismo , Animales , Procesamiento Proteico-Postraduccional , Glucógeno Fosforilasa/metabolismo , Glicosilación , Glucógeno Fosforilasa de Forma Muscular/metabolismo , Enfermedad de Alzheimer/metabolismo
2.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628557

RESUMEN

Glial cells participate actively in the early cognitive decline in Alzheimer's disease (AD) pathology. In fact, recent studies have found molecular and functional abnormalities in astrocytes and microglia in both animal models and brains of patients suffering from this pathology. In this regard, reactive gliosis intimately associated with amyloid plaques has become a pathological hallmark of AD. A recent study from our laboratory reports that astrocyte reactivity is caused by a direct interaction between amyloid beta (Aß) oligomers and integrin ß1. Here, we have generated four recombinant peptides including the extracellular domain of integrin ß1, and evaluated their capacity both to bind in vitro to Aß oligomers and to prevent in vivo Aß oligomer-induced gliosis and endoplasmic reticulum stress. We have identified the minimal region of integrin ß1 that binds to Aß oligomers. This region is called signal peptide and corresponds to the first 20 amino acids of the integrin ß1 N-terminal domain. This recombinant integrin ß1 signal peptide prevented Aß oligomer-induced ROS generation in primary astrocyte cultures. Furthermore, we carried out intrahippocampal injection in adult mice of recombinant integrin ß1 signal peptide combined with or without Aß oligomers and we evaluated by immunohistochemistry both astrogliosis and microgliosis as well as endoplasmic reticulum stress. The results show that recombinant integrin ß1 signal peptide precluded both astrogliosis and microgliosis and endoplasmic reticulum stress mediated by Aß oligomers in vivo. We have developed a molecular tool that blocks the activation of the molecular cascade that mediates gliosis via Aß oligomer/integrin ß1 signaling.


Asunto(s)
Péptidos beta-Amiloides , Gliosis , Integrina beta1 , Señales de Clasificación de Proteína , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Integrina beta1/metabolismo , Ratones
3.
Int J Mol Sci ; 23(9)2022 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-35563042

RESUMEN

Glycogen storage disease type V (GSDV, McArdle disease) is a rare genetic myopathy caused by deficiency of the muscle isoform of glycogen phosphorylase (PYGM). This results in a block in the use of muscle glycogen as an energetic substrate, with subsequent exercise intolerance. The pathobiology of GSDV is still not fully understood, especially with regard to some features such as persistent muscle damage (i.e., even without prior exercise). We aimed at identifying potential muscle protein biomarkers of GSDV by analyzing the muscle proteome and the molecular networks associated with muscle dysfunction in these patients. Muscle biopsies from eight patients and eight healthy controls showing none of the features of McArdle disease, such as frequent contractures and persistent muscle damage, were studied by quantitative protein expression using isobaric tags for relative and absolute quantitation (iTRAQ) followed by artificial neuronal networks (ANNs) and topology analysis. Protein candidate validation was performed by Western blot. Several proteins predominantly involved in the process of muscle contraction and/or calcium homeostasis, such as myosin, sarcoplasmic/endoplasmic reticulum calcium ATPase 1, tropomyosin alpha-1 chain, troponin isoforms, and alpha-actinin-3, showed significantly lower expression levels in the muscle of GSDV patients. These proteins could be potential biomarkers of the persistent muscle damage in the absence of prior exertion reported in GSDV patients. Further studies are needed to elucidate the molecular mechanisms by which PYGM controls the expression of these proteins.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo V , Proteoma , Biomarcadores/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo V/genética , Humanos , Músculo Esquelético/metabolismo , Isoformas de Proteínas/metabolismo , Proteoma/metabolismo
4.
Exerc Immunol Rev ; 27: 125-141, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33965896

RESUMEN

BACKGROUND: Regular exercise, particularly moderate-intensity continuous training (MICT), can improve immune function. Natural killer (NK) cells, a subset of lymphocytes that react to infections, are the most responsive innate immune cells to exercise, but the mechanisms underlying this are poorly understood. A type of exercise training that is gaining popularity in recent years is high-intensity interval training (HIIT), but how it affects NK cells is largely unknown. In fact, intense exercise has been traditionally viewed as a potential stressor to immune homeostasis. The purpose of this study was to determine in healthy, previously untrained adults (N=8 [3 male; 40±6 years]) the effects of an intervention consisting of 4-week MICT followed by 4-week HIIT on NK cells as compared with a pre-training (baseline) state. METHODS: Participants were studied at three time points: baseline, mid-intervention (after MICT), and post-intervention (after HIIT). Main assessments included cytotoxicity assays, flow-cytometry analysis of NK cell surface markers, and interrogation of the cellular proteome using a systems biology approach. RESULTS: A significant time effect was found for NK cell cytotoxicity (p<0.001), which was increased ~10-fold at both midand post-intervention versus baseline. No significant intervention effect was found for NK surface receptor expression, except for CXCR3 determined as mean fluorescence intensity (p=0.044, although with no significant differences in post hoc pairwise comparisons). The proteins showing a higher differential expression (Log2 fold-change > 10 and false discovery rate [FDR] q-value < 0.001) were COP9 signalosome subunit 3 (COPS3), DnaJ heat shock protein family member B11 (DNAJB11), histidyl-TRNA synthetase 1 (HARS), NIMA related kinase 9 (NEK9), nucleoporin 88 (NUP88), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), regulator of chromosome condensation 2 (RCC2), TAO kinase 3 (TAOK3), transducin beta like 2 (TBL2), and ring finger protein 40 (RNF40). All were upregulated at mid-intervention compared with baseline, with the exception of HARS, which was downregulated. Four enriched pathways (FDR p<25%) were found: two related to transmembrane transport and cellular composition (downregulated at mid-intervention vs baseline), and two related to oxidation- reduction reactions (regulated at post-intervention versus baseline). CONCLUSION: A progressive exercise intervention of MICT followed by HIIT induces a remarkable improvement in NK function compared with the untrained state, although at the mechanistic level the pathways involved seem to differ over time during the intervention.


Asunto(s)
Entrenamiento de Intervalos de Alta Intensidad , Células Asesinas Naturales/inmunología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteómica , Biología de Sistemas
5.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445683

RESUMEN

Neurodegenerative diseases are characterized by the progressive loss of specific subsets of neurons [...].


Asunto(s)
Comunicación Celular/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Transducción de Señal/fisiología , Animales , Humanos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo
6.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922618

RESUMEN

Small guanosine triphosphatases (GTPases) of the Rab and Arf families are key regulators of vesicle formation and membrane trafficking. Membrane transport plays an important role in the central nervous system. In this regard, neurons require a constant flow of membranes for the correct distribution of receptors, for the precise composition of proteins and organelles in dendrites and axons, for the continuous exocytosis/endocytosis of synaptic vesicles and for the elimination of dysfunctional proteins. Thus, it is not surprising that Rab and Arf GTPases have been associated with neurodegenerative diseases such as Alzheimer's and Parkinson's. Both pathologies share characteristics such as the presence of protein aggregates and/or the fragmentation of the Golgi apparatus, hallmarks that have been related to both Rab and Arf GTPases functions. Despite their relationship with neurodegenerative disorders, very few studies have focused on the role of these GTPases in the pathogenesis of neurodegeneration. In this review, we summarize their importance in the onset and progression of Alzheimer's and Parkinson's diseases, as well as their emergence as potential therapeutical targets for neurodegeneration.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Enfermedades Neurodegenerativas/patología , Proteínas de Unión al GTP rab/metabolismo , Animales , Exocitosis , Humanos , Enfermedades Neurodegenerativas/metabolismo , Transporte de Proteínas , Transducción de Señal
7.
Med Res Rev ; 40(4): 1315-1334, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32017179

RESUMEN

Human mesenchymal stromal cells (hMSCs) are emerging as one of the most important cell types in advanced therapies and regenerative medicine due to their great therapeutic potential. The development of hMSC-based products focuses on the use of hMSCs as biological active substances, and they are considered medicinal products by the primary health agencies worldwide. Due to their regulatory status, the development of hMSC-based products is regulated by specific criteria that range from the design phase, nonclinical studies, clinical studies, to the final registration and approval. Patients should only be administered hMSC-based products within the framework of a clinical trial or after the product has obtained marketing authorization; in both cases, authorization by health authorities is usually required. Considering the above, this paper describes the current general regulatory requirements for hMSC-based products, by jurisdiction, to be implemented throughout their entire development process. These measures may provide support for researchers from both public and private entities and academia to optimize the development of these products and their subsequent marketing, thereby improving access to them by patients.


Asunto(s)
Internacionalidad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Control Social Formal , Investigación Biomédica Traslacional , Humanos , Mercadotecnía
8.
J Biol Chem ; 294(12): 4345-4358, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30647127

RESUMEN

We recently uncovered a regulatory pathway of the muscle isoform of glycogen phosphorylase (PYGM) that plays an important role in regulating immune function in T cells. Here, using various enzymatic, pulldown, and immunoprecipitation assays, we describe signaling cross-talk between the small GTPases RAS and RAP1A, member of RAS oncogene family (RAP1) in human Kit 225 lymphoid cells, which, in turn, is regulated by the epidermal growth factor receptor (EGFR). We found that this communication bridge is essential for glycogen phosphorylase (PYG) activation through the canonical pathway because this enzyme is inactive in the absence of adenylyl cyclase type 6 (ADCY6). PYG activation required stimulation of both exchange protein directly activated by cAMP 2 (EPAC2) and RAP1 via RAS and ADCY6 phosphorylation, with the latter being mediated by Raf-1 proto-oncogene, Ser/Thr kinase (RAF1). Consistent with this model, PYG activation was EGFR-dependent and may be initiated by the constitutively active form of RAS. Consequently, PYG activation in Kit 225 T cells could be blocked with specific inhibitors of RAS, EPAC, RAP1, RAF1, ADCY6, and cAMP-dependent protein kinase. Our results establish a new paradigm for the mechanism of PYG activation, which depends on the type of receptor involved.


Asunto(s)
Receptores ErbB/fisiología , GTP Fosfohidrolasas/metabolismo , Glucógeno Fosforilasa/metabolismo , Linfocitos T/enzimología , Animales , Línea Celular , Activación Enzimática , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Fosforilación , Proto-Oncogenes Mas , Transducción de Señal , Proteínas de Unión al GTP rap1/metabolismo
9.
J Physiol ; 596(6): 1035-1061, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29315579

RESUMEN

KEY POINTS: Although they are unable to utilize muscle glycogen, McArdle mice adapt favourably to an individualized moderate-intensity endurance exercise training regime. Yet, they fail to reach the performance capacity of healthy mice with normal glycogen availability. There is a remarkable difference in the protein networks involved in muscle tissue adaptations to endurance exercise training in mice with and without glycogen availability. Indeed, endurance exercise training promoted the expression of only three proteins common to both McArdle and wild-type mice: LIMCH1, PARP1 and TIGD4. In turn, trained McArdle mice presented strong expression of mitogen-activated protein kinase 12 (MAPK12). ABSTRACT: McArdle's disease is an inborn disorder of skeletal muscle glycogen metabolism that results in blockade of glycogen breakdown due to mutations in the myophosphorylase gene. We recently developed a mouse model carrying the homozygous p.R50X common human mutation (McArdle mouse), facilitating the study of how glycogen availability affects muscle molecular adaptations to endurance exercise training. Using quantitative differential analysis by liquid chromatography with tandem mass spectrometry, we analysed the quadriceps muscle proteome of 16-week-old McArdle (n = 5) and wild-type (WT) (n = 4) mice previously subjected to 8 weeks' moderate-intensity treadmill training or to an equivalent control (no training) period. Protein networks enriched within the differentially expressed proteins with training in WT and McArdle mice were assessed by hypergeometric enrichment analysis. Whereas endurance exercise training improved the estimated maximal aerobic capacity of both WT and McArdle mice as compared with controls, it was ∼50% lower than normal in McArdle mice before and after training. We found a remarkable difference in the protein networks involved in muscle tissue adaptations induced by endurance exercise training with and without glycogen availability, and training induced the expression of only three proteins common to McArdle and WT mice: LIM and calponin homology domains-containing protein 1 (LIMCH1), poly (ADP-ribose) polymerase 1 (PARP1 - although the training effect was more marked in McArdle mice), and tigger transposable element derived 4 (TIGD4). Trained McArdle mice presented strong expression of mitogen-activated protein kinase 12 (MAPK12). Through an in-depth proteomic analysis, we provide mechanistic insight into how glycogen availability affects muscle protein signalling adaptations to endurance exercise training.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo V/fisiopatología , Glucógeno/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiología , Condicionamiento Físico Animal , Proteómica/métodos , Animales , Tolerancia al Ejercicio , Enfermedad del Almacenamiento de Glucógeno Tipo V/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mapas de Interacción de Proteínas
11.
J Biol Chem ; 290(14): 9171-82, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25694429

RESUMEN

Recently, we have reported that the active form of Rac 1 GTPase binds to the glycogen phosphorylase muscle isoform (PYGM) and modulates its enzymatic activity leading to T cell proliferation. In the lymphoid system, Rac 1 and in general other small GTPases of the Rho family participate in the signaling cascades that are activated after engagement of the T cell antigen receptor. However, little is known about the IL-2-dependent Rac 1 activator molecules. For the first time, a signaling pathway leading to the activation of Rac 1/PYGM in response to IL-2-stimulated T cell proliferation is described. More specifically, αPIX, a known guanine nucleotide exchange factor for the small GTPases of the Rho family, preferentially Rac 1, mediates PYGM activation in Kit 225 T cells stimulated with IL-2. Using directed mutagenesis, phosphorylation of αPIX Rho-GEF serines 225 and 488 is required for activation of the Rac 1/PYGM pathway. IL-2-stimulated serine phosphorylation was corroborated in Kit 225 T cells cultures. A parallel pharmacological and genetic approach identified PKCθ as the serine/threonine kinase responsible for αPIX serine phosphorylation. The phosphorylated state of αPIX was required to activate first Rac 1 and subsequently PYGM. These results demonstrate that the IL-2 receptor activation, among other early events, leads to activation of PKCθ. To activate Rac 1 and consequently PYGM, PKCθ phosphorylates αPIX in T cells. The biological significance of this PKCθ/αPIX/Rac 1 GTPase/PYGM signaling pathway seems to be the control of different cellular responses such as migration and proliferation.


Asunto(s)
Glucógeno Fosforilasa/metabolismo , Interleucina-12/farmacología , Linfocitos T/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Secuencia de Bases , Línea Celular , Proliferación Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Cartilla de ADN , Activación Enzimática , Humanos , Reacción en Cadena de la Polimerasa , Factores de Intercambio de Guanina Nucleótido Rho/fisiología , Linfocitos T/enzimología , Linfocitos T/metabolismo
12.
Chromosome Res ; 23(2): 285-98, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25645734

RESUMEN

Down syndrome is a common birth defect caused by trisomy of chromosome 21. Chromosomes occupy distinct territories in interphase nuclei, and their distribution within the nuclear space is nonrandom. In humans with Down syndrome, two chromosomes 21 frequently localize proximal to one another and distant from the third chromosome. Here, we investigated the nuclear organization of DYRK1A and SOD1, two genes mapping to chromosome 21 that greatly contribute to the pathology. We found that DYRK1A conserves its central positioning between normal and trisomic cells, whereas SOD1 adopts more peripheral distribution in trisomic cells. We also found that the relative position of these genes with respect to each other varies among the different copies of chromosome territories 21 within a cell, and that this distinct distribution is associated with differences in their expression levels. All together, our results may explain, at least in part, the difference in the expression level of these two genes implicated in the pathogenesis of Down syndrome.


Asunto(s)
Núcleo Celular/genética , Síndrome de Down/genética , Expresión Génica , Interfase/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Alelos , Línea Celular , Cromosomas Humanos Par 21/genética , Sitios Genéticos , Humanos , Hibridación Fluorescente in Situ , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Quinasas DyrK
13.
J Biol Chem ; 287(15): 11878-90, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22337875

RESUMEN

Small GTPases of the Rho family have been implicated in important cellular processes such as cell migration and adhesion, protein secretion, and/or gene transcription. In the lymphoid system, these GTPases participate in the signaling cascades that are activated after engagement of antigen receptors. However, little is known about the role that Rho GTPases play in IL-2-mediated responses. Here, we show that IL-2 induces Rac1 activation in Kit 225 T cells. We identified by mass spectrometry the muscle isoform of glycogen phosphorylase (PYGM) as a novel Rac1 effector molecule in IL-2-stimulated cells. The interaction between the active form of Rac1 (Rac1-GTP) and PYGM was established directly through a domain comprising amino acids 191-270 of PYGM that exhibits significant homology with the Rac binding domain of PAK1. The integrity of this region was crucial for PYGM activation. Importantly, IL-2-dependent cellular proliferation was inhibited upon blocking both the activation of Rac1 and the activity of PYGM. These results reveal a new role for Rac1 in cell signaling, showing that this GTPase triggers T cell proliferation upon IL-2 stimulation by associating with PYGM and modulating its enzymatic activity.


Asunto(s)
Proliferación Celular , Activación Enzimática , Glucógeno Fosforilasa de Forma Muscular/metabolismo , Interleucina-2/fisiología , Linfocitos T/fisiología , Proteína de Unión al GTP rac1/fisiología , Secuencia de Aminoácidos , Línea Celular , Expresión Génica , Glucógeno Fosforilasa de Forma Muscular/química , Glucógeno Fosforilasa de Forma Muscular/genética , Humanos , Interleucina-2/metabolismo , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Mapeo Peptídico , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal , Linfocitos T/enzimología , Proteína de Unión al GTP rac1/metabolismo
14.
Cells ; 12(14)2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37508540

RESUMEN

Protein O-GlcNAcylation has been associated with neurodegenerative diseases such as Alzheimer's disease (AD). The O-GlcNAcylation of the Amyloid Precursor Protein (APP) regulates both the trafficking and the processing of the APP through the amyloidogenic pathway, resulting in the release and aggregation of the Aß1-42 peptide. Microglia clears Aß aggregates and dead cells to maintain brain homeostasis. Here, using LC-MS/MS, we revealed that the Aß1-42 oligomers modify the microglia O-GlcNAcome. We identified 55 proteins, focusing our research on Galectin-1 protein since it is a very versatile protein from a functional point of view. Combining biochemical with genetic approaches, we demonstrated that Aß1-42 oligomers specifically target Galectin-1S8 O-GlcNAcylation via OGT. In addition to this, the Gal-1-O-GlcNAcylated form, in turn, controls human microglia migration. Given the importance of microglia migration in the progression of AD, this study reports the relationship between the Aß1-42 oligomers and Serine 8-O-GlcNAcylation of Galectin-1 to drive microglial migration.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Galectina 1/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo
15.
Cell Death Dis ; 13(3): 253, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-35306512

RESUMEN

Amyloid beta (Aß)-mediated synapse dysfunction is an early event in Alzheimer's disease (AD) pathogenesis and previous studies suggest that NMDA receptor (NMDAR) dysregulation may contribute to these pathological effects. Although Aß peptides impair NMDAR expression and activity, the mechanisms mediating these alterations in the early stages of AD are unclear. Here, we observed that NMDAR subunit NR2B and PSD-95 levels were aberrantly upregulated and correlated with Aß42 load in human postsynaptic fractions of the prefrontal cortex in early stages of AD patients, as well as in the hippocampus of 3xTg-AD mice. Importantly, NR2B and PSD95 dysregulation was revealed by an increased expression of both proteins in Aß-injected mouse hippocampi. In cultured neurons, Aß oligomers increased the NR2B-containing NMDAR density in neuronal membranes and the NMDA-induced intracellular Ca2+ increase, in addition to colocalization in dendrites of NR2B subunit and PSD95. Mechanistically, Aß oligomers required integrin ß1 to promote synaptic location and function of NR2B-containing NMDARs and PSD95 by phosphorylation through classic PKCs. These results provide evidence that Aß oligomers modify the contribution of NR2B to NMDAR composition and function in the early stages of AD through an integrin ß1 and PKC-dependent pathway. These data reveal a novel role of Aß oligomers in synaptic dysfunction that may be relevant to early-stage AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Proteína Quinasa C/metabolismo , Receptores de N-Metil-D-Aspartato , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Hipocampo/metabolismo , Humanos , Integrina beta1/metabolismo , Ratones , N-Metilaspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
16.
Nat Cell Biol ; 5(7): 633-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12819788

RESUMEN

Cyclic AMP (cAMP) is produced by activation of Gs protein-coupled receptors and regulates many physiological processes through activation of protein kinase A (PKA). However, a large body of evidence indicates that cAMP also regulates specific cellular functions through PKA-independent pathways. Here, we show that a small GTPase of the Rho family, Rac, is regulated by cAMP in a PKA-independent manner. We also show that Rac activation results from activation of Rap1 through the cAMP guanine nucleotide-exchange factor (GEF) Epac1. Activation of the Gs-coupled serotonin 5-HT(4) receptor initiates this signalling cascade in various cell types. Furthermore, we demonstrate that crosstalk between the Ras and Rho GTPase families is involved in cAMP-dependent processing of amyloid precursor protein (APP), a key protein in Alzheimer's disease. Indeed, Epac1 regulates secretion of the non-amyloidogenic soluble form of APP (sAPPalpha) through Rap1 and Rac. Our data identify an unsuspected connection between two families of small GTPases and imply that Rac can function downstream of cAMP/Epac1/Rap1 in a novel signal transduction secretory pathway.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Precursor de Proteína beta-Amiloide/metabolismo , Corteza Cerebral/enzimología , Neuronas/enzimología , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/biosíntesis , Animales , Células CHO , Corteza Cerebral/fisiopatología , Cricetinae , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Receptores de Serotonina/metabolismo , Receptores de Serotonina 5-HT4 , Transducción de Señal/genética , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rap1/genética
17.
Small GTPases ; 12(2): 106-113, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-31512989

RESUMEN

Small GTPases, together with their regulatory and effector molecules, are key intermediaries in the complex signalling pathways that control almost all cellular processes, working as molecular switches to transduce extracellular cues into cellular responses that drive vital functions, such as intracellular transport, biomolecule synthesis, gene activation and cell survival. How all of these networks are linked to metabolic pathways is a subject of intensive study. Because any response to cellular action requires some form of energy input, elucidating how cells coordinate the signals that lead to a tangible response involving metabolism is central to understand cellular activities. In this review, we summarize recent advances in our understanding of the molecular basis of the crosstalk between small GTPases of the Ras superfamily, specifically Rac1 and Ras/Rap1, and glycogen phosphorylase in T lymphocytes. Abbreviations: ADCY: adenylyl cyclase; ADCY6: adenylyl cyclase 6; BCR: B cell receptor; cAMP: 3',5'-cyclic adenosine monophosphate; CRIB: Cdc42/Rac binding domain; DLPFC: dysfunction of the dorsolateral prefrontal cortex; EGFR: epidermal growth factor receptor; Epac2: exchange protein directly activated by cAMP; GDP: guanodine-5'-diphosphate; GPCRs: G protein-coupled receptors; GTP: guanodin-5'-triphosphate; IL2: interleukin 2; IL2-R: interleukin 2 receptor; JAK: janus kinases; MAPK: mitogen-activated protein kinase; O-GlcNAc: O-glycosylation; PAK1: p21 activated kinase 1; PI3K: phosphatidylinositol 3-kinase; PK: phosphorylase kinase; PKA: cAMP-dependent protein kinase A; PKCθ: protein kinase Cθ; PLCγ: phospholipase Cγ; Src: proto-oncogene tyrosine-protein kinase c; STAT: signal transducer and activator of transcription proteins.


Asunto(s)
Proteínas de Unión al GTP Monoméricas
18.
Neuropharmacology ; 185: 108081, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32407924

RESUMEN

When Alzheimer's disease (AD) disease-modifying therapies will be available, global healthcare systems will be challenged by a large-scale demand for clinical and biological screening. Validation and qualification of globally accessible, minimally-invasive, and time-, cost-saving blood-based biomarkers need to be advanced. Novel pathophysiological mechanisms (and related candidate biomarkers) - including neuroinflammation pathways (TREM2 and YKL-40), axonal degeneration (neurofilament light chain protein), synaptic dysfunction (neurogranin, synaptotagmin, α-synuclein, and SNAP-25) - may be integrated into an expanding pathophysiological and biomarker matrix and, ultimately, integrated into a comprehensive blood-based liquid biopsy, aligned with the evolving ATN + classification system and the precision medicine paradigm. Liquid biopsy-based diagnostic and therapeutic algorithms are increasingly employed in Oncology disease-modifying therapies and medical practice, showing an enormous potential for AD and other brain diseases as well. For AD and other neurodegenerative diseases, newly identified aberrant molecular pathways have been identified as suitable therapeutic targets and are currently investigated by academia/industry-led R&D programs, including the nerve-growth factor pathway in basal forebrain cholinergic neurons, the sigma1 receptor, and the GTPases of the Rho family. Evidence for a clinical long-term effect on cognitive function and brain health span of cholinergic compounds, drug candidates for repositioning programs, and non-pharmacological multidomain interventions (nutrition, cognitive training, and physical activity) is developing as well. Ultimately, novel pharmacological paradigms, such as quantitative systems pharmacology-based integrative/explorative approaches, are gaining momentum to optimize drug discovery and accomplish effective pathway-based strategies for precision medicine. This article is part of the special issue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/tratamiento farmacológico , Descubrimiento de Drogas/tendencias , Líquido Intracelular/efectos de los fármacos , Farmacología Clínica/tendencias , Biología de Sistemas/tendencias , Enfermedad de Alzheimer/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/metabolismo , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Reposicionamiento de Medicamentos/tendencias , Predicción , Humanos , Líquido Intracelular/metabolismo , Biopsia Líquida/métodos , Biopsia Líquida/tendencias , Glicoproteínas de Membrana/metabolismo , Farmacología Clínica/métodos , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Biología de Sistemas/métodos
19.
Artículo en Inglés | MEDLINE | ID: mdl-32116638

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder that leads to impaired memory and cognitive deficits. Spine loss as well as changes in spine morphology correlates with cognitive impairment in this neurological disorder. Many studies in animal models and ex vivo cultures indicate that amyloid ß-peptide (Aß) oligomers induce synaptic damage early during the progression of the disease. Here, in order to determine the events that initiate synaptic alterations, we acutely applied oligomeric Aß to primary hippocampal neurons and an ex vivo model of organotypic hippocampal cultures from a mouse after targeted expression of EGFP to allow high-resolution imaging and algorithm-based evaluation of spine changes. Dendritic spines were classified as thin, stubby or mushroom, based on morphology. In vivo, time-lapse imaging showed that the three spine types were relatively stable, although their stability significantly decreased after treatment with Aß oligomers. Unexpectedly, we observed that the density of total dendritic spines increased in organotypic hippocampal slices treated with Aß compared to control cultures. Specifically, the fraction of stubby spines significantly increased, while mushroom and thin spines remained unaltered. Pharmacological tools revealed that acute Aß oligomers induced spine changes through mechanisms involving CaMKII and integrin ß1 activities. Additionally, analysis of dendritic complexity based on a 3D reconstruction of the whole neuron morphology showed an increase in the apical dendrite length and branching points in CA1 organotypic hippocampal slices treated with Aß. In contrast to spines, the morphological changes were affected by integrin ß1 but not by CaMKII inhibition. Altogether, these data indicate that the Aß oligomers exhibit early dual effects by acutely enhancing dendritic complexity and spine density.

20.
Aging (Albany NY) ; 12(4): 3502-3515, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32100723

RESUMEN

Healthy aging depends on a complex gene-environment network that is ultimately reflected in the expression of different proteins. We aimed to perform a comparative analysis of the plasma proteome of healthy centenarians (n=9, 5 women, age range 100-103 years) with a notably preserved ambulatory capacity (as a paradigm of 'successful' aging), and control individuals who died from a major age-related disease before the expected life expectancy (n=9, 5 women, age range: 67-81 years), and while having impaired ambulatory capacity (as a paradigm of 'unsuccessful' aging). We found that the expression of 49 proteins and 86 pathways differed between the two groups. Overall, healthy centenarians presented with distinct expression of proteins/pathways that reflect a healthy immune function, including a lower pro-inflammatory status (less 'inflammaging' and autoimmunity) and a preserved humoral immune response (increased B cell-mediated immune response). Compared with controls, healthy centenarians also presented with a higher expression of proteins involved in angiogenesis and related to enhanced intercellular junctions, as well as a lower expression of proteins involved in cardiovascular abnormalities. The identification of these proteins/pathways might provide new insights into the biological mechanisms underlying the paradigm of healthy aging.


Asunto(s)
Envejecimiento Saludable/metabolismo , Proteoma/metabolismo , Caminata/fisiología , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Femenino , Estado de Salud , Humanos , Inflamación/metabolismo , Masculino , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA