Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochem Soc Trans ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38661212

ABSTRACT

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.

2.
Cells ; 12(14)2023 07 17.
Article in English | MEDLINE | ID: mdl-37508540

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/metabolism , Microglia/metabolism , Galectin 1/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism
3.
Int J Mol Sci ; 23(10)2022 May 20.
Article in English | MEDLINE | ID: mdl-35628557

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides , Gliosis , Integrin beta1 , Protein Sorting Signals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Humans , Integrin beta1/metabolism , Mice
4.
Exerc Immunol Rev ; 27: 125-141, 2021.
Article in English | MEDLINE | ID: mdl-33965896

ABSTRACT

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.


Subject(s)
High-Intensity Interval Training , Killer Cells, Natural/immunology , Adult , Female , Humans , Male , Middle Aged , Proteomics , Systems Biology
5.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922618

ABSTRACT

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.


Subject(s)
ADP-Ribosylation Factors/metabolism , Neurodegenerative Diseases/pathology , rab GTP-Binding Proteins/metabolism , Animals , Exocytosis , Humans , Neurodegenerative Diseases/metabolism , Protein Transport , Signal Transduction
6.
Neuropharmacology ; 185: 108081, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32407924

ABSTRACT

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'.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/drug therapy , Drug Discovery/trends , Intracellular Fluid/drug effects , Pharmacology, Clinical/trends , Systems Biology/trends , Alzheimer Disease/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Drug Discovery/methods , Drug Repositioning/methods , Drug Repositioning/trends , Forecasting , Humans , Intracellular Fluid/metabolism , Liquid Biopsy/methods , Liquid Biopsy/trends , Membrane Glycoproteins/metabolism , Pharmacology, Clinical/methods , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Systems Biology/methods
7.
Small GTPases ; 12(2): 106-113, 2021 03.
Article in English | MEDLINE | ID: mdl-31512989

ABSTRACT

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.


Subject(s)
Monomeric GTP-Binding Proteins
8.
Int J Mol Sci ; 21(17)2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32878220

ABSTRACT

Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer's disease (AD) and Parkinson's disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases.


Subject(s)
Monomeric GTP-Binding Proteins/metabolism , Neurodegenerative Diseases/physiopathology , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Humans , Signal Transduction
9.
Aging (Albany NY) ; 12(4): 3502-3515, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32100723

ABSTRACT

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.


Subject(s)
Healthy Aging/metabolism , Proteome/metabolism , Walking/physiology , Aged , Aged, 80 and over , Aging/metabolism , Female , Health Status , Humans , Inflammation/metabolism , Male , Proteomics
10.
Int J Mol Sci ; 20(23)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31775340

ABSTRACT

McArdle disease, also known as glycogen storage disease type V (GSDV), is characterized by exercise intolerance, the second wind phenomenon, and high serum creatine kinase activity. Here, we recapitulate PYGM mutations in the population responsible for this disease. Traditionally, McArdle disease has been considered a metabolic myopathy caused by the lack of expression of the muscle isoform of the glycogen phosphorylase (PYGM). However, recent findings challenge this view, since it has been shown that PYGM is present in other tissues than the skeletal muscle. We review the latest studies about the molecular mechanism involved in glycogen phosphorylase activity regulation. Further, we summarize the expression and functional significance of PYGM in other tissues than skeletal muscle both in health and McArdle disease. Furthermore, we examine the different animal models that have served as the knowledge base for better understanding of McArdle disease. Finally, we give an overview of the latest state-of-the-art clinical trials currently being carried out and present an updated view of the current therapies.


Subject(s)
Glycogen Phosphorylase/metabolism , Glycogen Storage Disease Type V/pathology , Muscle, Skeletal/pathology , Animals , Glycogen Storage Disease Type V/enzymology , Humans , Muscle, Skeletal/enzymology
12.
J Biol Chem ; 294(12): 4345-4358, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30647127

ABSTRACT

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.


Subject(s)
ErbB Receptors/physiology , GTP Phosphohydrolases/metabolism , Glycogen Phosphorylase/metabolism , T-Lymphocytes/enzymology , Animals , Cell Line , Enzyme Activation , Guanine Nucleotide Exchange Factors/metabolism , Humans , Phosphorylation , Proto-Oncogene Mas , Signal Transduction , rap1 GTP-Binding Proteins/metabolism
13.
Front Mol Neurosci ; 11: 333, 2018.
Article in English | MEDLINE | ID: mdl-30271323

ABSTRACT

Oligodendrocytes are highly vulnerable to glutamate excitotoxicity, a central mechanism involved in tissue damage in Multiple Sclerosis (MS). Sustained activation of AMPA receptors in rat oligodendrocytes induces cytosolic calcium overload, mitochondrial depolarization, increase of reactive oxygen species, and activation of intracelular pathways resulting in apoptotic cell death. Although many signals driven by excitotoxicity have been identified, some of the key players are still under investigation. Casein kinase 2 (CK2) is a serine/threonine kinase, constitutively expressed in all eukaryotic tissues, involved in cell proliferation, malignant transformation and apoptosis. In this study, we identify CK2 as a critical regulator of oligodendrocytic death pathways and elucidate its role as a signal inductor following excitotoxic insults. We provide evidence that CK2 activity is up-regulated in AMPA-treated oligodendrocytes and CK2 inhibition significantly diminished AMPA receptor-induced oligodendroglial death. In addition, we analyzed mitogen-activated protein kinase (MAPK) signaling after excitotoxic insult. We observed that AMPA receptor activation induced a rapid increase in c-Jun N-terminal kinase (JNK) and p38 phosphorylation that was reduced after CK2 inhibition. Moreover, blocking their phosphorylation, we enhanced oligodendrocyte survival after excitotoxic insult. Finally, we observed that the tumor suppressor p53 is activated during AMPA receptor-induced cell death and, interestingly, down-regulated by JNK or CK2 inhibition. Together, these data indicate that the increase in CK2 activity induced by excitotoxic insults regulates MAPKs, triggers p53 activation and mediates subsequent oligodendroglial loss. Therefore, targeting CK2 may be a useful strategy to prevent oligodendrocyte death in MS and other diseases involving central nervous system (CNS) white matter.

14.
Oncotarget ; 9(67): 32775-32794, 2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30214684

ABSTRACT

Hepatocellular carcinoma develops as a multistep process, in which cell cycle deregulation is a central feature, resulting in unscheduled proliferation. The PLAGL1 gene encodes a homonym zinc finger protein that is involved in cell-proliferation control. We determined the genomic profile and the transcription and expression level of PLAGL1, simultaneously with that of its molecular partners p53, PPARγ and p21, in cell-lines derived from patients with liver cancer, during in vitro cell growth. Our investigations revealed that genomic and epigenetic changes of PLAGL1 are also present in hepatoma cell-lines. Transcription of PLAGL1 in tumor cells is significantly lower than in normal fibroblasts, but no significant differences in terms of protein expression were detected between these two cell-types, indicating that there is not a direct relationship between the gene transcriptional activity and protein expression. RT-PCR analyses on normal fibroblasts, used as control, also showed that PLAGL1 and p53 genes transcription occurs as an apparent orchestrated process during normal cells proliferation, which gets disturbed in cancer cells. Furthermore, abnormal trafficking of the PLAGL1 protein may occur in hepatocarcinogenesis.

15.
J Physiol ; 596(6): 1035-1061, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29315579

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Glycogen Storage Disease Type V/physiopathology , Glycogen/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Proteomics/methods , Animals , Exercise Tolerance , Glycogen Storage Disease Type V/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Interaction Maps
16.
Aging Cell ; 15(6): 1140-1152, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27709751

ABSTRACT

Astrogliosis is a hallmark of Alzheimer's disease (AD) and may constitute a primary pathogenic component of that disorder. Elucidation of signaling cascades inducing astrogliosis should help characterizing the function of astrocytes and identifying novel molecular targets to modulate AD progression. Here, we describe a novel mechanism by which soluble amyloid-ß modulates ß1-integrin activity and triggers NADPH oxidase (NOX)-dependent astrogliosis in vitro and in vivo. Amyloid-ß oligomers activate a PI3K/classical PKC/Rac1/NOX pathway which is initiated by ß1-integrin in cultured astrocytes. This mechanism promotes ß1-integrin maturation, upregulation of NOX2 and of the glial fibrillary acidic protein (GFAP) in astrocytes in vitro and in hippocampal astrocytes in vivo. Notably, immunochemical analysis of the hippocampi of a triple-transgenic AD mouse model shows increased levels of GFAP, NOX2, and ß1-integrin in reactive astrocytes which correlates with the amyloid ß-oligomer load. Finally, analysis of these proteins in postmortem frontal cortex from different stages of AD (II to V/VI) and matched controls confirmed elevated expression of NOX2 and ß1-integrin in that cortical region and specifically in reactive astrocytes, which was most prominent at advanced AD stages. Importantly, protein levels of NOX2 and ß1-integrin were significantly associated with increased amyloid-ß load in human samples. These data strongly suggest that astrogliosis in AD is caused by direct interaction of amyloid ß oligomers with ß1-integrin which in turn leads to enhancing ß1-integrin and NOX2 activity via NOX-dependent mechanisms. These observations may be relevant to AD pathophysiology.

17.
Cell Signal ; 28(11): 1713-24, 2016 11.
Article in English | MEDLINE | ID: mdl-27519475

ABSTRACT

Recently, we have reported that the IL-2-stimulated T cells activate PKCθ in order to phosphorylate the serine residues of αPIX-RhoGEF, and to switch on the Rac1/PYGM pathway resulting in T cell migration and proliferation. However, the molecular mechanism connecting the activated IL-2-R with the PKCθ/αPIX/Rac1/PYGM pathway is still unknown. In this study, the use of a combined pharmacological and genetic approach identified Lck, a Src family member, as the tyrosine kinase phosphorylating PLCγ leading to Rac1 and PYGM activation in the IL-2-stimulated Kit 225 T cells via the PKCθ/αPIX pathway. The PLCγ tyrosine phosphorylation was required to activate first PKCθ, and then αPIX and Rac1/PYGM. The results presented here delineate a novel signalling pathway ranking equally in importance to the three major pathways controlled by the IL-2-R, i.e. PI3K, Ras/MAPK and JAK/STAT pathways. The overall evidence strongly indicates that the central biological role of the novel IL-2-R/Lck/PLCγ/PKCθ/αPIX/Rac1/PYGM signalling pathway is directly related to the control of fundamental cellular processes such as T cell migration and proliferation.


Subject(s)
Cell Movement , Glycogen Phosphorylase/metabolism , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phospholipase C gamma/metabolism , T-Lymphocytes/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemotaxis/drug effects , Enzyme Activation/drug effects , Humans , Models, Biological , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Kinase C/metabolism , Receptors, Interleukin-2/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects , src-Family Kinases/metabolism
18.
FEBS J ; 283(4): 647-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663827

ABSTRACT

Small GTPases of the Ras superfamily are capable of activating E2F-dependent transcription leading to cell proliferation, but the molecular mechanisms are poorly understood. In this study, using immortalized chicken DT40 B cell lines to investigate the role of the Vav/Rac signalling cascade on B cell proliferation, it is shown that the proliferative response triggered by B cell receptor activation is dramatically reduced in the absence of Vav3 expression. Analysis of this proliferative defect shows that in the absence of Vav3 expression, retinoblastoma protein (RB) phosphorylation and the subsequent E2F activation do not take place. By combining pharmacological and genetic approaches, phosphatidylinositol-3-kinase and phospholipase Cγ2 (PLCγ2) were identified as the key regulatory signalling molecules upstream of the Vav3/Rac pathway leading to RB phosphorylation and E2F transcription factor activation. Additionally, vav3(-/-) and plcγ2(-/-) DT40 B cells were not able to activate the RB-E2F complex wild-type phenotype when these genetically modified cells were transfected with constitutively active forms of RhoA or Cdc42. However, when these knockout cells were transfected with different constitutively active versions of PLCγ, Vav or Rac1, not only activation of the RB-E2F complex wild-type phenotype was recovered but also the cellular proliferation. Furthermore, by evaluating the effect of two known effector mutants of Rac1 (Rac1(Q61L/F37A) and Rac1(Q61L/Y40C) ), the RB-E2F complex activation dependency on p21-activated kinase (PAK) and protein kinase Cε (PKCε) activities was established, being independent of both actin cytoskeleton reorganization and Ras activity. These results suggest that PAK1 and PKCε may be potential therapeutic targets to stop uncontrolled B cell proliferation mediated by the Vav/Rac pathway.


Subject(s)
B-Lymphocytes/metabolism , E2F Transcription Factors/metabolism , Retinoblastoma Protein/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , B-Lymphocytes/cytology , Cell Line , Cell Proliferation , Chickens , Phosphorylation , Signal Transduction
19.
J Biol Chem ; 290(14): 9171-82, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25694429

ABSTRACT

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.


Subject(s)
Glycogen Phosphorylase/metabolism , Interleukin-12/pharmacology , T-Lymphocytes/drug effects , rac1 GTP-Binding Protein/metabolism , Base Sequence , Cell Line , Cell Proliferation/drug effects , Chemotaxis, Leukocyte/drug effects , DNA Primers , Enzyme Activation , Humans , Polymerase Chain Reaction , Rho Guanine Nucleotide Exchange Factors/physiology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...