Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
PLoS One ; 14(3): e0213508, 2019.
Article in English | MEDLINE | ID: mdl-30870435

ABSTRACT

During experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis associated with blood-brain barrier (BBB) disruption, oligodendrocyte precursor cells (OPCs) overexpress proteoglycan nerve/glial antigen 2 (NG2), proliferate, and make contacts with the microvessel wall. To explore whether OPCs may actually be recruited within the neurovascular unit (NVU), de facto intervening in its cellular and molecular composition, we quantified by immunoconfocal morphometry the presence of OPCs in contact with brain microvessels, during postnatal cerebral cortex vascularization at postnatal day 6, in wild-type (WT) and NG2 knock-out (NG2KO) mice, and in the cortex of adult naïve and EAE-affected WT and NG2KO mice. As observed in WT mice during postnatal development, a higher number of juxtavascular and perivascular OPCs was revealed in adult WT mice during EAE compared to adult naïve WT mice. In EAE-affected mice, OPCs were mostly associated with microvessels that showed altered claudin-5 and occludin tight junction (TJ) staining patterns and barrier leakage. In contrast, EAE-affected NG2KO mice, which did not show any significant increase in vessel-associated OPCs, seemed to retain better preserved TJs and BBB integrity. As expected, absence of NG2, in both OPCs and pericytes, led to a reduced content of vessel basal lamina molecules, laminin, collagen VI, and collagen IV. In addition, analysis of the major ligand/receptor systems known to promote OPC proliferation and migration indicated that vascular endothelial growth factor A (VEGF-A), platelet-derived growth factor-AA (PDGF-AA), and the transforming growth factor-ß (TGF-ß) were the molecules most likely involved in proliferation and recruitment of vascular OPCs during EAE. These results were confirmed by real time-PCR that showed Fgf2, Pdgfa and Tgfb expression on isolated cerebral cortex microvessels and by dual RNAscope-immunohistochemistry/in situ hybridization (IHC/ISH), which detected Vegfa and Vegfr2 transcripts on cerebral cortex sections. Overall, this study suggests that vascular OPCs, in virtue of their developmental arrangement and response to neuroinflammation and growth factors, could be integrated among the classical NVU cell components. Moreover, the synchronized activation of vascular OPCs and pericytes during both BBB development and dysfunction, points to NG2 as a key regulator of vascular interactions.


Subject(s)
Antigens/biosynthesis , Blood-Brain Barrier/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Microvessels/metabolism , Oligodendroglia/metabolism , Proteoglycans/biosynthesis , Stem Cells/metabolism , Animals , Antigens/genetics , Blood-Brain Barrier/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Claudin-5/genetics , Claudin-5/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Knockout , Microvessels/pathology , Oligodendroglia/pathology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Proteoglycans/genetics , Stem Cells/pathology , Tight Junctions/genetics , Tight Junctions/metabolism , Tight Junctions/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Trials ; 17(1): 549, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27855714

ABSTRACT

BACKGROUND: In this multi-centre randomized controlled trial (RCT) we compared modified mentalisation-based treatment (MBT-ED) to specialist supportive clinical management (SSCM-ED) in patients with eating disorders (EDs) and borderline personality disorder symptoms (BPD). This group of patients presents complex challenges to clinical services, and a treatment which addresses their multiple problems has the potential to improve outcome. MBT has been shown to be effective in improving outcome in patients with BPD, but its use has not been reported in ED. METHODS: Sixty-eight eligible participants were randomised to MBT-ED or SSCM-ED. The primary outcome measure was the global score on the Eating Disorder Examination. Secondary outcomes included measures of BPD symptoms (the Zanarini Rating Scale for Borderline Personality Disorder), general psychiatric state, quality of life and service utilisation. Participants were assessed at baseline and at 6, 12 and 18 months after randomisation. Analysis was performed using linear mixed models. RESULTS: Only 15 participants (22 %) completed the 18 month follow-up. Early drop-out occurred significantly more in the SSCM-ED group. Drop-out did not vary with treatment model later in therapy and was sometimes attributed to participants moving away. There was higher drop--out amongst smokers and those with higher neuroticism scores. 47.1 % of participants in the MBT-ED arm and 37.1 % in the SSCM-ED arm attended at least 50 % of therapy sessions offered. Amongst those remaining in the trial, at 12 and 18 months MBT-ED was associated with a greater reduction in Shape Concern and Weight Concern in the Eating Disorder Examination compared to SSCM-ED. At 6, 12 and 18 months there was a decline of ED and BPD symptoms in both groups combined. Ten participants were reported as having had adverse events during the trial, mostly self-harm, and there was one death, attributed as 'unexplained' by the coroner. CONCLUSIONS: The high drop-out rate made interpretation of the results difficult. Greater involvement of research staff in clinical management might have improved compliance with both therapy and research assessment. MBT-ED may have had an impact on core body image psychopathology. TRIAL REGISTRATION: Current Controlled Trials: ISRCTN51304415 . Registered on 19 April 2011.


Subject(s)
Borderline Personality Disorder/therapy , Feeding and Eating Disorders/therapy , Theory of Mind , Adult , Female , Humans , Male , Patient Compliance , Patient Selection , Single-Blind Method
3.
Acta Neuropathol ; 132(1): 23-42, 2016 07.
Article in English | MEDLINE | ID: mdl-27026411

ABSTRACT

In adult CNS, nerve/glial-antigen 2 (NG2) is expressed by oligodendrocyte progenitor cells (OPCs) and is an early marker of pericyte activation in pathological conditions. NG2 could, therefore, play a role in experimental autoimmune encephalomyelitis (EAE), a disease associated with increased blood-brain barrier (BBB) permeability, inflammatory infiltrates, and CNS damage. We induced EAE in NG2 knock-out (NG2KO) mice and used laser confocal microscopy immunofluorescence and morphometry to dissect the effect of NG2 KO on CNS pathology. NG2KO mice developed milder EAE than their wild-type (WT) counterparts, with less intense neuropathology associated with a significant improvement in BBB stability. In contrast to WT mice, OPC numbers did not change in NG2KO mice during EAE. Through FACS and confocal microscopy, we found that NG2 was also expressed by immune cells, including T cells, macrophages, and dendritic cells (DCs). Assessment of recall T cell responses to the encephalitogen by proliferation assays and ELISA showed that, while WT and NG2KO T cells proliferated equally to the encephalitogenic peptide MOG35-55, NG2KO T cells were skewed towards a Th2-type response. Because DCs could be responsible for this effect, we assessed their expression of IL-12 by PCR and intracellular FACS. IL-12-expressing CD11c+ cells were significantly decreased in MOG35-55-primed NG2KO lymph node cells. Importantly, in WT mice, the proportion of IL-12-expressing cells was significantly lower in CD11c+ NG2- cells than in CD11c+ NG2+ cells. To assess the relevance of NG2 at immune system and CNS levels, we induced EAE in bone-marrow chimeric mice, generated with WT recipients of NG2KO bone-marrow cells and vice versa. Regardless of their original phenotype, mice receiving NG2KO bone marrow developed milder EAE than those receiving WT bone marrow. Our data suggest that NG2 plays a role in EAE not only at CNS/BBB level, but also at immune response level, impacting on DC activation and thereby their stimulation of reactive T cells, through controlling IL-12 expression.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
4.
Aging (Albany NY) ; 8(1): 34-49, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26761436

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells that critically influence decisions about immune activation or tolerance. Impaired DC function is at the core of common chronic disorders and contributes to reduce immunocompetence during aging. Knowledge on the mechanisms regulating DC generation and function is necessary to understand the immune system and to prevent disease and immunosenescence. Here we show that the sirtuin Sirt6, which was previously linked to healthspan promotion, stimulates the development of myeloid, conventional DCs (cDCs). Sirt6-knockout (Sirt6KO) mice exhibit low frequencies of bone marrow cDC precursors and low yields of bone marrow-derived cDCs compared to wild-type (WT) animals. Sirt6KO cDCs express lower levels of class II MHC, of costimulatory molecules, and of the chemokine receptor CCR7, and are less immunostimulatory compared to WT cDCs. Similar effects in terms of differentiation and immunostimulatory capacity were observed in human monocyte-derived DCs in response to SIRT6 inhibition. Finally, while Sirt6KO cDCs show an overall reduction in their ability to produce IL-12, TNF-α and IL-6 secretion varies dependent on the stimulus, being reduced in response to CpG, but increased in response to other Toll-like receptor ligands. In conclusion, Sirt6 plays a crucial role in cDC differentiation and function and reduced Sirt6 activity may contribute to immunosenescence.


Subject(s)
Cell Differentiation , Dendritic Cells/enzymology , Immunosenescence , Sirtuins/metabolism , Animals , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Genotype , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Histone Deacetylase Inhibitors/pharmacology , Immunosenescence/drug effects , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Sirtuins/antagonists & inhibitors , Sirtuins/deficiency , Sirtuins/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
5.
Acta Neuropathol ; 130(2): 279-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25920452

ABSTRACT

Dimethyl fumarate (DMF), recently approved as an oral immunomodulatory treatment for relapsing-remitting multiple sclerosis (MS), metabolizes to monomethyl fumarate (MMF) which crosses the blood-brain barrier and has demonstrated neuroprotective effects in experimental studies. We postulated that MMF exerts neuroprotective effects through modulation of microglia activation, a critical component of the neuroinflammatory cascade that occurs in neurodegenerative diseases such as MS. To ascertain our hypothesis and define the mechanistic pathways involved in the modulating effect of fumarates, we used real-time PCR and biochemical assays to assess changes in the molecular and functional phenotype of microglia, quantitative Western blotting to monitor activation of postulated pathway components, and ex vivo whole-cell patch clamp recording of excitatory post-synaptic currents in corticostriatal slices from mice with experimental autoimmune encephalomyelitis (EAE), a model for MS, to study synaptic transmission. We show that exposure to MMF switches the molecular and functional phenotype of activated microglia from classically activated, pro-inflammatory type to alternatively activated, neuroprotective one, through activation of the hydroxycarboxylic acid receptor 2 (HCAR2). We validate a downstream pathway mediated through the AMPK-Sirt1 axis resulting in deacetylation, and thereby inhibition, of NF-κB and, consequently, of secretion of pro-inflammatory molecules. We demonstrate through ex vivo monitoring of spontaneous glutamate-mediated excitatory post-synaptic currents of single neurons in corticostriatal slices from EAE mice that the neuroprotective effect of DMF was exerted on neurons at pre-synaptic terminals by modulating glutamate release. By exposing control slices to untreated and MMF-treated activated microglia, we confirm the modulating effect of MMF on microglia function and, thereby, its indirect neuroprotective effect at post-synaptic level. These findings, whereby DMF-induced activation of a new HCAR2-dependent pathway on microglia leads to the modulation of neuroinflammation and restores synaptic alterations occurring in EAE, represent a possible novel mechanism of action for DMF in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Fumarates/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Synapses/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Brain/drug effects , Brain/physiopathology , Cell Line , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Excitatory Postsynaptic Potentials/drug effects , Female , Glutamic Acid/metabolism , Mice, Inbred C57BL , Microglia/physiology , NF-kappa B/metabolism , Signal Transduction/physiology , Sirtuin 1/metabolism , Synapses/physiology , Tissue Culture Techniques
6.
J Neuropathol Exp Neurol ; 71(10): 840-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23001217

ABSTRACT

The pathophysiology of cerebral cortical lesions in multiple sclerosis (MS) is not understood. We investigated cerebral cortex microvessels during immune-mediated demyelination in the MS model chronic murine experimental autoimmune encephalomyelitis (EAE) by immunolocalization of the endothelial cell tight junction (TJ) integral proteins claudin-5 and occludin, a structural protein of caveolae, caveolin-1, and the blood-brain barrier-specific endothelial transporter, Glut 1. In EAE-affected mice, there were areas of extensive subpial demyelination and well-demarcated lesions that extended to deeper cortical layers. Activation of microglia and absence of perivascular inflammatory infiltrates were common in these areas. Microvascular endothelial cells showed increased expression of caveolin-1 and a coincident loss of both claudin-5 and occludin normal junctional staining patterns. At a very early disease stage, claudin-5 molecules tended to cluster and form vacuoles that were also Glut 1 positive; the initially preserved occludin pattern became diffusely cytoplasmic at more advanced stages. Possible internalization of claudin-5 on TJ dismantling was suggested by its coexpression with the autophagosomal marker MAP1LC3A. Loss of TJ integrity was confirmed by fluorescein isothiocyanate-dextran experiments that showed leakage of the tracer into the perivascular neuropil. These observations indicate that, in the cerebral cortex of EAE-affected mice, there is a microvascular disease that differentially targets claudin-5 and occludin during ongoing demyelination despite only minimal inflammation.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Animals , Blood-Brain Barrier/pathology , Caveolin 1/metabolism , Cerebral Cortex/pathology , Claudin-5/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/metabolism , Mice , Mice, Inbred C57BL , Occludin/metabolism
7.
Mol Med ; 18: 794-804, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22481270

ABSTRACT

Despite some advances in the understanding of amyotrophic lateral sclerosis (ALS) pathogenesis, significant achievements in treating this disease are still lacking. Mesenchymal stromal (stem) cells (MSCs) have been shown to be effective in several models of neurological disease. To determine the effects of the intravenous injection of MSCs in an ALS mouse model during the symptomatic stage of disease, MSCs (1 × 106) were intravenously injected in mice expressing human superoxide dismutase 1 (SOD1) carrying the G93A mutation (SOD1/G93A) presenting with experimental ALS. Survival, motor abilities, histology, oxidative stress markers and [³H]D-aspartate release in the spinal cord were investigated. MSC injection in SOD1/G93A mice improved survival and motor functions compared with saline-injected controls. Injected MSCs scantly home to the central nervous system and poorly engraft. We observed a reduced accumulation of ubiquitin agglomerates and of activated astrocytes and microglia in the spinal cord of MSC-treated SOD1/G93A mice, with no changes in the number of choline acetyltransferase- and glutamate transporter type 1-positive cells. MSC administration turned around the upregulation of metallothionein mRNA expression and of the activity of the antioxidant enzyme glutathione S-transferase, both associated with disease progression. Last, we observed that MSCs reverted both spontaneous and stimulus-evoked neuronal release of [³H]D-aspartate, a marker of endogenous glutamate, which is upregulated in SOD1/G93A mice. These findings suggest that intravenous administration of MSCs significantly improves the clinical outcome and pathological scores of mutant SOD1/G93A mice, thus providing the rationale for their exploitation for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Mesenchymal Stem Cell Transplantation , Motor Activity , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/mortality , Animals , Aspartic Acid/metabolism , Cell Movement , Central Nervous System/metabolism , Central Nervous System/physiopathology , Disease Progression , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Oxidative Stress , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
8.
Stem Cell Res Ther ; 3(1): 3, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22277374

ABSTRACT

Stem cells are currently seen as a treatment for tissue regeneration in neurological diseases such as multiple sclerosis, anticipating that they integrate and differentiate into neural cells. Mesenchymal stem cells (MSCs), a subset of adult progenitor cells, differentiate into cells of the mesodermal lineage but also, under certain experimental circumstances, into cells of the neuronal and glial lineage. Their clinical development, however, has been significantly boosted by the demonstration that MSCs display significant therapeutic plasticity mainly occurring through bystander mechanisms. These features have been exploited in the effective treatment of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis where the inhibition of the autoimmune response resulted in a significant amelioration of disease and decrease of demyelination, immune infiltrates and axonal loss. Surprisingly, these effects do not require MSCs to engraft in the central nervous system but depend on the cells' ability to inhibit pathogenic immune responses both in the periphery and inside the central nervous system and to release neuroprotective and pro-oligodendrogenic molecules favoring tissue repair. These results paved the road for the utilization of MSCs for the treatment of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , Central Nervous System/immunology , Drug Administration Routes , Forkhead Transcription Factors/metabolism
9.
Proc Natl Acad Sci U S A ; 108(42): 17384-9, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21960443

ABSTRACT

Dendritic cells (DC) are highly specialized antigen-presenting cells characterized by the ability to prime T-cell responses. Mesenchymal stem cells (MSC) are adult stromal progenitor cells displaying immunomodulatory activities including inhibition of DC maturation in vitro. However, the specific impact of MSC on DC functions, upon in vivo administration, has never been elucidated. Here we show that murine MSC impair Toll-like receptor-4 induced activation of DC resulting in the inhibition of cytokines secretion, down-regulation of molecules involved in the migration to the lymph nodes, antigen presentation to CD4(+) T cells, and cross-presentation to CD8(+) T cells. These effects are associated with the inhibition of phosphorylation of intracellular mitogen-activated protein kinases. Intravenous administration of MSC decreased the number of CCR7 and CD49dß1 expressing CFSE-labeled DC in the draining lymph nodes and hindered local antigen priming of DO11.10 ovalbumin-specific CD4(+) T cells. Upon labeling of DC with technetium-99m hexamethylpropylene amine oxime to follow their in vivo biodistribution, we demonstrated that intravenous injection of MSC blocks, almost instantaneously, the migration of subcutaneously administered ovalbumin-pulsed DC to the draining lymph nodes. These findings indicate that MSC significantly affect DC ability to prime T cells in vivo because of their inability to home to the draining lymph nodes and further confirm MSC potentiality as therapy for immune-mediated diseases.


Subject(s)
Dendritic Cells/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Coculture Techniques , Cytokines/genetics , Dendritic Cells/cytology , Dendritic Cells/physiology , Dendritic Cells/transplantation , Gene Expression , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
10.
Curr Stem Cell Res Ther ; 6(1): 69-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20955153

ABSTRACT

The lack of therapies fostering remyelination and regeneration of the neural network deranged by the autoimmune attack occurring in multiple sclerosis (MS), is raising great expectations about stem cells therapies for tissue repair. Mesenchymal stem cells (MSCs) have been proposed as a possible treatment for MS due to the reported capacity of transdifferentiation into neural cells and their ability at modulating immune responses. However, recent studies have demonstrated that many other functional properties are likely to play a role in the therapeutic plasticity of MSCs, including anti-apoptotic, trophic and anti-oxidant effects. These features are mostly based on the paracrine release of soluble molecules, often dictated by local environmental cues. Based on the modest evidence of long-term engraftment and the striking clinical effects that are observed immediately after MSCs administration in the experimental model of MS, we do not favor a major role for transdifferentiation as an important mechanism involved in the therapeutic effect of MSCs.


Subject(s)
Cell Transdifferentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Multiple Sclerosis/therapy , Animals , Humans , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology
11.
Psychiatry Res ; 187(3): 401-8, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21095017

ABSTRACT

This exploratory study assessed attachment and personality in anorexic women, non-affected siblings, and healthy controls, examining correlations with psychopathology. Thirty-eight anorexic subjects (31 females), thirty-one siblings (22 females), and fifty controls (35 females) participated. Personality development characteristics were assessed using the Attachment Style Questionnaire (ASQ), Temperament and Character Inventory (TCI), Eating Disorders Inventory (EDI-2), Symptom Checklist-90 (SCL-90), and other inventories for clinical assessment of EDs. Both anorexic probands and their siblings described lower maternal care and higher maternal overprotection than did controls. Healthy siblings were more similar to controls, but had lower scores than either controls or affected siblings on preoccupation with relationships (P<0.005) and higher scores than controls on self-transcendence (P<0.015) and obsessive-compulsive traits (P<0.025). Logistic regression indicated that need for approval, persistence, resourcefulness, self-transcendence, state anger, pursuit of thinness, interpersonal distrust, social insecurity, and binge eating differentiated anorexic probands from siblings. The need for approval was related to several psychopathological characteristics. Low preoccupation with relationships, low need for approval, and high self-transcendence may have protected siblings from family and environmental stressors. High need for approval was independently related to psychopathological traits in anorexic siblings. Implications for prevention and treatment are discussed.


Subject(s)
Anorexia Nervosa/physiopathology , Anorexia Nervosa/psychology , Personality Development , Siblings , Adolescent , Adult , Analysis of Variance , Anorexia Nervosa/genetics , Body Mass Index , Female , Humans , Logistic Models , Personality Inventory , Psychiatric Status Rating Scales , Psychopathology , Surveys and Questionnaires , Young Adult
12.
J Neurochem ; 115(2): 343-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20649849

ABSTRACT

We have investigated the spontaneous and the depolarisation-induced release of [(3)H]D-aspartate ([(3)H]D-ASP), a non-metabolisable analogue of glutamate, in spinal cord slices, synaptosomes and gliosomes from mice with experimental autoimmune encephalomyelitis (EAE) at 13, 21 and 55 days post-immunisation (d.p.i.), representing onset, peak and chronic phases of the pathology. At 13 and 21 d.p.i., the KCl-evoked, calcium-dependent overflow of [(3)H]D-ASP in spinal cord slices was significantly lower (30-40%), whereas at 55 d.p.i. it was significantly higher (30%), than that elicited in matched controls. When the release was measured from spinal cord synaptosomes and gliosomes in superfusion, a different picture emerged. The spontaneous and the KCl(15 mM)-induced release of [(3)H]D-ASP were significantly increased both in synaptosomes (17% and 45%, respectively) and gliosomes (26% and 25%, respectively) at 21, but not at 13, d.p.i. At 55 d.p.i., the KCl-induced [(3)H]D-ASP release was significantly increased (40%) only in synaptosomes. Finally, uptake of [(3)H]D-ASP was markedly (50-60%) increased in spinal cord synaptosomes, but not in gliosomes, obtained from EAE mice at 21 d.p.i., whereas no differences could be detected at 13 d.p.i. Our data indicate that glutamatergic neurotransmission is altered in the spinal cord of EAE mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Glutamic Acid/metabolism , Spinal Cord/metabolism , Animals , Aspartic Acid/metabolism , Disease Models, Animal , Female , Glycoproteins/immunology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Peptide Fragments/immunology , Potassium Chloride/pharmacology , Spinal Cord/drug effects , Spinal Cord/pathology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tritium/metabolism
13.
PLoS One ; 4(11): e7897, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19936064

ABSTRACT

Nicotinamide phosphoribosyltransferase (Nampt) inhibitors such as FK866 are potent inhibitors of NAD(+) synthesis that show promise for the treatment of different forms of cancer. Based on Nampt upregulation in activated T lymphocytes and on preliminary reports of lymphopenia in FK866 treated patients, we have investigated FK866 for its capacity to interfere with T lymphocyte function and survival. Intracellular pyridine nucleotides, ATP, mitochondrial function, viability, proliferation, activation markers and cytokine secretion were assessed in resting and in activated human T lymphocytes. In addition, we used experimental autoimmune encephalomyelitis (EAE) as a model of T-cell mediated autoimmune disease to assess FK866 efficacy in vivo. We show that activated, but not resting, T lymphocytes undergo massive NAD(+) depletion upon FK866-mediated Nampt inhibition. As a consequence, impaired proliferation, reduced IFN-gamma and TNF-alpha production, and finally autophagic cell demise result. We demonstrate that upregulation of the NAD(+)-degrading enzyme poly-(ADP-ribose)-polymerase (PARP) by activated T cells enhances their susceptibility to NAD(+) depletion. In addition, we relate defective IFN-gamma and TNF-alpha production in response to FK866 to impaired Sirt6 activity. Finally, we show that FK866 strikingly reduces the neurological damage and the clinical manifestations of EAE. In conclusion, Nampt inhibitors (and possibly Sirt6 inhibitors) could be used to modulate T cell-mediated immune responses and thereby be beneficial in immune-mediated disorders.


Subject(s)
Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Lymphocyte Activation , Myelin Sheath/chemistry , NAD/chemistry , Nicotinamide Phosphoribosyltransferase/metabolism , T-Lymphocytes/metabolism , Acrylamides/pharmacology , Adenosine Triphosphate/chemistry , Animals , Autophagy , Cell Proliferation , Female , Humans , Interferon-gamma/metabolism , Jurkat Cells , Membrane Potentials , Mice , Mice, Inbred C57BL , NAD/metabolism , Piperidines/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
J Neurochem ; 110(5): 1674-84, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19619133

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), an animal model for human multiple sclerosis, is characterized by demyelination, inflammation and neurodegeneration of CNS in which free radicals play a role. Recently, the efficacy of murine mesenchimal stem cells (MSCs) as treatment of EAE induced in mice by the encephalitogenic peptide MOG(35-55) was demonstrated. The present study analyzed some markers of oxidative stress, inflammation/degeneration and apoptosis such as metallothioneins (MTs), antioxidant enzymes (superoxide dismutase, catalase and glutathione-S-transferase), poly(ADP-ribose) polymerase-1 and p53 during EAE progression and following MSC treatment. Expression of the three brain MT isoforms increased significantly in EAE mice compared with healthy controls, but while expression of MT-1 and MT-3 increased along EAE course, MT-2 was up-regulated at the onset, but returned to levels similar to those of controls in chronic phase. The changes in the transcription and activity of the antioxidant enzymes and in expression of poly(ADP-ribose) polymerase-1 and p53 showed the same kinetics observed for MT-1 and MT-3 during EAE. Interestingly, i.v. administration of MSCs reduced the EAE-induced increases in levels/activities of all these proteins. These results support an antioxidant and neuroprotective activity for MSCs that was also confirmed in vitro on neuroblastoma cells exposed to an oxidative insult.


Subject(s)
Antioxidants/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Animals , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Humans , Mesenchymal Stem Cells/cytology , Metallothionein/metabolism , Metallothionein 3 , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...