Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 4: e406, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24984193

ABSTRACT

We here present data on immune gene expression of chemokines, chemokine receptors, cytokines and regulatory T-cell (T-reg) markers in chronic patients suffering from either schizophrenia (SCZ, N=20) or bipolar disorder (BD=20) compared with healthy controls (HCs, N=20). We extracted RNA from peripheral blood mononuclear cells and performed real-time (RT)-PCR to measure mRNA levels of chemokines, chemokine receptors, cytokines and T-reg markers. All the analyses were Bonferroni-corrected. The classical monocyte activation (M1) markers il6, ccl3 were significantly increased in BD as compared with both HC and SCZ patients (P=0.03 and P=0.002; P=0.024 and P=0.021, respectively), whereas markers of alternative (M2) monocyte activation ccl1, ccl22 and il10 were coherently decreased (controls: P=0.01, P=0.001 and P=0.09; SCZ subjects: P=0.02, P=0.05 and P=0.011, respectively). Concerning T-cell markers, BD patients had compared with HC downregulated ccr5 (P=0.02) and upregulated il4 (P=0.04) and compared with both healthy and SCZ individuals downregulated ccl2 (P=0.006 and P=0.003) and tgfß (P=0.004 and P=0.007, respectively). No significant associations were found between any immune gene expression and clinical variables (prior hospitalizations, Brief Psychiatric Rating Scale, medications' dosages and lifetime administration). Although some markers are expressed by different immune cell types, these findings suggest a coherent increased M1/decrease M2 signature in the peripheral blood of BD patients with potential Th1/Th2 shift. In contrast, all the explored immune marker levels were preserved in SCZ. Further larger studies are needed to investigate the relevance of inflammatory response in BD, trying to correlate it to psychopathology, treatment and outcome measures and, possibly, to brain connectivity.


Subject(s)
Bipolar Disorder/immunology , Cytokines/immunology , Monocytes/immunology , Schizophrenia/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adult , Biomarkers/blood , Chronic Disease , Female , Gene Expression , Humans , Male , Middle Aged , RNA, Messenger
2.
Cell Death Differ ; 21(4): 582-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24336048

ABSTRACT

Alzheimer's disease (AD) is characterized by extracellular amyloid-ß (Aß) deposition, which activates microglia, induces neuroinflammation and drives neurodegeneration. Recent evidence indicates that soluble pre-fibrillar Aß species, rather than insoluble fibrils, are the most toxic forms of Aß. Preventing soluble Aß formation represents, therefore, a major goal in AD. We investigated whether microvesicles (MVs) released extracellularly by reactive microglia may contribute to AD degeneration. We found that production of myeloid MVs, likely of microglial origin, is strikingly high in AD patients and in subjects with mild cognitive impairment and that AD MVs are toxic for cultured neurons. The mechanism responsible for MV neurotoxicity was defined in vitro using MVs produced by primary microglia. We demonstrated that neurotoxicity of MVs results from (i) the capability of MV lipids to promote formation of soluble Aß species from extracellular insoluble aggregates and (ii) from the presence of neurotoxic Aß forms trafficked to MVs after Aß internalization into microglia. MV neurotoxicity was neutralized by the Aß-interacting protein PrP and anti-Aß antibodies, which prevented binding to neurons of neurotoxic soluble Aß species. This study identifies microglia-derived MVs as a novel mechanism by which microglia participate in AD degeneration, and suggest new therapeutic strategies for the treatment of the disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Microglia/metabolism , Neurons/drug effects , Peptide Fragments/toxicity , Transport Vesicles/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Female , Humans , Interleukin-1beta/metabolism , Male , Microglia/drug effects , Neurons/cytology , Neurons/metabolism , Peptide Fragments/chemistry , PrPC Proteins/metabolism , Rats , Solubility , Transport Vesicles/chemistry , Tumor Necrosis Factor-alpha/metabolism
3.
Gene Ther ; 20(5): 487-96, 2013 May.
Article in English | MEDLINE | ID: mdl-22855093

ABSTRACT

Interleukin-25 (IL-25) is the only anti-inflammatory cytokine of the IL-17 family, and it has been shown to be efficacious in inhibiting neuroinflammation. Known for its effects on cells of the adaptive immune system, it has been more recently described to be effective also on cells of the innate immune system, namely macrophages. We used a lentiviral-mediated gene therapy approach to deliver IL-25 to the central nervous system (CNS) in two mouse models of neuroinflammation, entorhinal cortex lesion and experimental autoimmune encephalomyelitis. In both, we found that IL-25 gene therapy was able to modulate CNS myeloid cells, either infiltrating macrophages or resident microglia, towards an anti-inflammatory, tissue-protective phenotype, as testified by the increase in markers such as Arginase-1 (Arg1), Mannose receptor 1 (CD206) and Chitinase 3-like 3 (Ym1). As a consequence, neuroinflammation was partly inhibited and the CNS protected from immune-mediated damage. To our knowledge, this is the first example of M2 shift (alternative activation) induced in vivo on CNS-resident myeloid cells by gene therapy, and may constitute a promising strategy to investigate the potential role of protective microglia in neurological disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Entorhinal Cortex , Genetic Therapy , Inflammation/therapy , Interleukin-17/genetics , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Humans , Inflammation/genetics , Interleukin-17/therapeutic use , Lentivirus/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/pathology , Microglia/transplantation , Myeloid Cells/metabolism , Myeloid Cells/pathology
4.
Mult Scler ; 17(11): 1301-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21677024

ABSTRACT

BACKGROUND: Inflammation triggers secondary neurodegeneration in multiple sclerosis (MS). OBJECTIVES: It is unclear whether classical anti-inflammatory cytokines have the potential to interfere with synaptic transmission and neuronal survival in MS. METHODS: Correlation analyses between cerebrospinal fluid (CSF) contents of anti-inflammatory cytokines and molecular, imaging, clinical, and neurophysiological measures of neuronal alterations were performed. RESULTS: Our data suggest that interleukin-13 (IL-13) plays a neuroprotective role in MS brains. We found, in fact, that the levels of IL-13 in the CSF of MS patients were correlated with the contents of amyloid-ß(1-42). Correlations were also found between IL-13 and imaging indexes of axonal and neuronal integrity, such as the retinal nerve fibre layer thickness and the macular volume evaluated by optical coherence tomography. Furthermore, the levels of IL-13 were related to better performance in the low-contrast acuity test and Multiple Sclerosis Functional Composite scoring. Finally, by means of transcranial magnetic stimulation, we have shown that GABAA-mediated cortical inhibition was more pronounced in patients with high IL-13 levels in the CSF, as expected for a neuroprotective, anti-excitotoxic effect. CONCLUSIONS: The present correlation study provides some evidence for the involvement of IL-13 in the modulation of neuronal integrity and synaptic function in patients with MS.


Subject(s)
Interleukin-13/cerebrospinal fluid , Motor Cortex/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Nerve Degeneration/immunology , Neurons/immunology , Adult , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Case-Control Studies , Chi-Square Distribution , Contrast Sensitivity , Disability Evaluation , Evoked Potentials, Motor , Female , Humans , Italy , Magnetic Resonance Imaging , Male , Middle Aged , Motor Cortex/pathology , Motor Cortex/physiopathology , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/pathology , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Nerve Degeneration/cerebrospinal fluid , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurons/pathology , Peptide Fragments/cerebrospinal fluid , Synaptic Transmission , Tomography, Optical Coherence , Transcranial Magnetic Stimulation , Young Adult
5.
J Neuroimmunol ; 209(1-2): 33-9, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19232440

ABSTRACT

Based on gene expression data, we tested the P8A-CCL2 variant of the chemokine CCL2, able to interfere with the chemotactic properties of the parental molecule, in relapsing-remitting (RR)-EAE SJL. Only preventive treatment significantly delayed disease onset in a dose dependent manner. P8A-CCL2 administration, however, decreased demyelination, axonal loss and number of CNS infiltrating T cells and macrophages. Immunological analysis revealed that P8A-CCL2 does not act on Ag-specific T cell proliferation and does not interfere with the differentiation of IFNgamma-releasing effectors T cells. These results suggest that the therapeutic mechanism of P8A-CCL2 may rely on interference with immune cell recruitment.


Subject(s)
Chemokine CCL2/pharmacology , Chemotaxis, Leukocyte/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Sheath/drug effects , Adult , Animals , Cell Proliferation/drug effects , Chemokine CCL2/chemical synthesis , Chemokine CCL2/therapeutic use , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interferon-gamma/metabolism , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myelin Sheath/immunology , Myelin Sheath/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Wallerian Degeneration/drug therapy , Wallerian Degeneration/immunology , Wallerian Degeneration/physiopathology
6.
Gene Ther ; 15(7): 504-15, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18239607

ABSTRACT

Central nervous system (CNS) delivery of anti-inflammatory cytokines, such as interleukin 4 (IL4), holds promise as treatment for multiple sclerosis (MS). We have previously shown that short-term herpes simplex virus type 1-mediated IL4 gene therapy is able to inhibit experimental autoimmune encephalomyelitis (EAE), an animal model of MS, in mice and non-human primates. Here, we show that a single administration of an IL4-expressing helper-dependent adenoviral vector (HD-Ad) into the cerebrospinal fluid (CSF) circulation of immunocompetent mice allows persistent transduction of neuroepithelial cells and long-term (up to 5 months) CNS transgene expression without toxicity. Mice affected by chronic and relapsing EAE display clinical and neurophysiological recovery from the disease once injected with the IL4-expressing HD-Ad vector. The therapeutic effect is due to the ability of IL4 to increase, in inflamed CNS areas, chemokines (CCL1, CCL17 and CCL22) capable of recruiting regulatory T cells (CD4+CD69-CD25+Foxp3+) with suppressant functions. CSF delivery of HD-Ad vectors expressing anti-inflammatory molecules might represent a valuable therapeutic option for CNS inflammatory disorders.


Subject(s)
Central Nervous System/immunology , Genetic Therapy/methods , Interleukin-4/genetics , Multiple Sclerosis/therapy , T-Lymphocytes, Regulatory/immunology , Adenoviridae/genetics , Animals , Central Nervous System/pathology , Chemokines/immunology , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Helper Viruses/genetics , Humans , Interleukin-4/analysis , Interleukin-4/immunology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic/methods
7.
Gene Ther ; 15(3): 233-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17960158

ABSTRACT

Inflammation and immune reaction, or pre-existing immunity towards commonly used viral vectors for gene therapy severely impair long-term gene expression in the central nervous system (CNS), impeding the possibility to repeat the therapeutic intervention. Here, we show that injection of a helper-dependent adenoviral (HD-Ad) vector by lumbar puncture into the cerebrospinal fluid (CSF) of non-human primates allows long-term (three months) infection of neuroepithelial cells, also in monkeys bearing a pre-existing anti-adenoviral immunity. Intrathecal injection of the HD-Ad vector was not associated with any sign of systemic or local toxicity, nor by signs of a CNS-specific immune reaction towards the HD-Ad vector. Injection of HD-Ad vectors into the CSF circulation may thus represent a valuable approach for CNS gene therapy allowing for long-term expression and re-administration.


Subject(s)
Adenoviridae/genetics , Cerebrospinal Fluid/virology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Helper Viruses/genetics , Parkinson Disease/therapy , Animals , Gene Expression , Genetic Engineering , Genetic Vectors/immunology , Interleukin-4/genetics , Macaca fascicularis , Male , Models, Animal , Neuroepithelial Cells/immunology , Neuroepithelial Cells/virology , Parkinson Disease/immunology , Spinal Puncture , Transduction, Genetic/methods
8.
Gene Ther ; 14(1): 93-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16929354

ABSTRACT

Primary proinflammatory cytokines, such as IL-1beta, play a crucial pathogenic role in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE), and may represent, therefore, a suitable therapeutic target. We have previously established the delivery of anti-inflammatory cytokine genes within the central nervous system (CNS), based on intracisternal (i.c.) injection of non-replicative HSV-1-derived vectors. Here we show the therapeutic efficacy of i.c. administration of an HSV-1-derived vector carrying the interleukin-1receptor antagonist (IL-1ra) gene, the physiological antagonist of the proinflammatory cytokine IL-1, in C57BL/6 mice affected by myelin oligodendrocyte glycoprotein-induced EAE. IL-1ra gene therapy is effective preventively, delaying EAE onset by almost 1 week (22.4+/-1.4 days post-immunization vs 15.9+/-2.1 days in control mice; P=0.0229 log-rank test), and decreasing disease severity. Amelioration of EAE course was associated with a reduced number of macrophages infiltrating the CNS and in a decreased level of proinflammatory cytokine mRNA in the CNS, suggesting an inhibitory activity of IL-1ra on effector cell recruitment, as antigen-specific peripheral T-cell activation and T-cell recruitment to the CNS is unaffected. Thus, local IL-1ra gene therapy may represent a therapeutic alternative for the inhibition of immune-mediated demyelination of the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , Interleukin 1 Receptor Antagonist Protein/genetics , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Cisterna Magna , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Genetic Vectors/genetics , Injections , Interferon-gamma/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myelin Proteins , Myelin-Associated Glycoprotein , Myelin-Oligodendrocyte Glycoprotein , RNA, Messenger/analysis , T-Lymphocytes/immunology , Time Factors , Tumor Necrosis Factor-alpha/genetics
9.
Gene Ther ; 8(16): 1207-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509953

ABSTRACT

The development of therapies aimed to promote remyelination is a major issue in chronic inflammatory demyelinating disorders of the central nervous system (CNS) such as multiple sclerosis (MS), where the permanent neurological impairment is due to the axonal loss resulting from recurrent episodes of immune-mediated demyelination. Here, we show that the intrathecal injection of a herpes simplex virus (HSV) type-1 replication-defective multigene vector, engineered with the human fibroblast growth factor (FGF)-II gene (TH:bFGF vector), was able to significantly revert in C57BL/6 mice the clinicopathological signs of chronic experimental autoimmune encephalomyelitis (EAE), the animal model of MS. The treatment with the TH:bFGF vector was initiated within 1 week after the clinical onset of EAE and was effective throughout the whole follow-up period (ie 60 days). The disease-ameliorating effect in FGF-II-treated mice was associated with: (1) CNS production of FGF-II from vector-infected cells which were exclusively located around the CSF space (ependymal, choroidal and leptomeningeal cells); (2) significant decrease (P < 0.01) of the number of myelinotoxic cells (T cells and macrophages) both in the CNS parenchyma and in the leptomeningeal space; and (3) significant increase (P < 0.01) of the number of oligodendrocyte precursors and of myelin-forming oligodendrocytes in areas of demyelination and axonal loss. Our results indicate that CNS gene therapy using HSV-1-derived vector coding for neurotrophic factors (ie FGF-II) is a safe and non-toxic approach that might represent a potential useful 'alternative' tool for the future treatment of immune-mediated demyelinating diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Fibroblast Growth Factor 2/genetics , Genetic Therapy/methods , Animals , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Vectors/administration & dosage , Injections, Spinal , Lymphocyte Count , Macrophages/immunology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/therapy , Oligodendroglia/pathology , Simplexvirus/genetics , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
10.
J Immunol ; 167(3): 1821-9, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466408

ABSTRACT

The exclusive detrimental role of proinflammatory cytokines in demyelinating diseases of the CNS, such as multiple sclerosis, is controversial. Here we show that the intrathecal delivery of an HSV-1-derived vector engineered with the mouse IFN-gamma gene leads to persistent (up to 4 wk) CNS production of IFN-gamma and inhibits the course of a chronic-progressive form of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by myelin oligodendrocyte glycoprotein (MOG)(35-55). Mice treated with the IFN-gamma-containing vector before EAE onset showed an earlier onset but a milder course of the disease compared with control mice treated with the empty vector. In addition, 83% of IFN-gamma-treated mice completely recovered within 25 days post immunization, whereas control mice did not recover up to 60 days post immunization. Mice treated with the IFN-gamma-containing vector within 1 wk after EAE onset partially recovered from the disease within 25 days after vector injection, whereas control mice worsened. Recovery from EAE in mice treated with IFN-gamma was associated with a significant increase of CNS-infiltrating lymphocytes undergoing apoptosis. During the recovery phase, the mRNA level of TNFR1 was also significantly increased in CNS-infiltrating cells from IFN-gamma-treated mice compared with controls. Our results further challenge the exclusive detrimental role of IFN-gamma in the CNS during EAE/multiple sclerosis, and indicate that CNS-confined inflammation may induce protective immunological countermechanisms leading to a faster clearance of encephalitogenic T cells by apoptosis, thus restoring the immune privilege of the CNS.


Subject(s)
Apoptosis/immunology , Brain/immunology , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-gamma/administration & dosage , Lymphocyte Subsets/pathology , Spinal Cord/immunology , Animals , Antigens, CD/biosynthesis , Apoptosis/genetics , Blood-Brain Barrier/genetics , Blood-Brain Barrier/immunology , Brain/metabolism , Brain/pathology , Cell Movement/genetics , Cerebral Ventricles/immunology , Cerebral Ventricles/virology , Chronic Disease , Cisterna Magna , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , Injections , Injections, Spinal , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Lymphocyte Count , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor, Type I , Spinal Cord/metabolism , Spinal Cord/pathology , Subarachnoid Space/immunology , Subarachnoid Space/virology , Virus Replication/genetics
11.
Hum Gene Ther ; 12(8): 905-20, 2001 May 20.
Article in English | MEDLINE | ID: mdl-11387056

ABSTRACT

Systemic administration of antiinflammatory molecules to patients affected by immune-mediated inflammatory demyelinating diseases of the central nervous system (CNS) has limited therapeutic efficacy due to the presence of the blood-brain barrier (BBB). We found that three of five rhesus monkeys injected intrathecally with a replication-defective herpes simplex virus (HSV) type 1-derived vector engineered with the human interleukin 4 (IL-4) gene were protected from an hyperacute and lethal form of experimental autoimmune encephalomyelitis induced by whole myelin. The intrathecally injected vector consistently diffused within the CNS via the cerebrospinal fluid and infected ependymal cells, which in turn sustained in situ production of IL-4 without overt immunological or toxic side effects. In EAE-protected monkeys, IL-4-gene therapy significantly decreased the number of brain as well as spinal cord inflammatory perivenular infiltrates and the extent of demyelination, necrosis, and axonal loss. The protective effect was associated with in situ downregulation of inflammatory mediators such as tumor necrosis factor alpha (TNF-alpha) and monocyte chemoattractant protein 1 (MCP-1), upregulation of transforming growth factor beta (TGF-beta), and preservation of BBB integrity. Our results indicate that intrathecal delivery of HSV-1-derived vectors containing antiinflammatory cytokine genes may play a major role in the future therapeutic armamentarium of inflammatory CNS-confined demyelinating diseases and, in particular, in the most fulminant forms where conventional therapeutic approaches have, so far, failed to achieve a satisfactory control of the disease evolution.


Subject(s)
Autoimmune Diseases/genetics , Central Nervous System/metabolism , Encephalomyelitis/genetics , Genetic Therapy/methods , Genetic Vectors , Herpesvirus 1, Human/genetics , Interleukin-4/genetics , Animals , Autoimmune Diseases/prevention & control , Blood-Brain Barrier , Brain/pathology , Cell Division , Chemokine CCL2/biosynthesis , Cytokines/biosynthesis , Down-Regulation , Encephalomyelitis/prevention & control , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-4/biosynthesis , Macaca mulatta , Magnetic Resonance Imaging , Male , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology , T-Lymphocytes/cytology , Time Factors , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
12.
Gene Ther ; 8(1): 13-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402297

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated inflammatory disease of the central nervous system (CNS) that might benefit from anti-inflammatory therapies. However, systemic delivery of anti-inflammatory drugs in MS patients has so far been disappointing, mostly due to the limited capacity of these molecules to enter the CNS. We injected into the cisterna magna (i.c.) of Biozzi AB/H mice affected by a relapsing-remitting form of experimental autoimmune encephalomyelitis (EAE), the animal model of MS, a non-replicative herpes simplex virus (HSV) type-1-derived vector containing the interleukin (IL)-4 gene (d120:LacZ:IL-4). CNS delivery of the d120:LacZ:IL-4 vector, after EAE onset, induced the in situ production of IL-4 by CNS-resident cells facing the cerebrospinal fluid (CSF) spaces and reduced by 47% (P < 0.02) the disease-related deaths. Compared with mice treated with the control d120:lacZ vector, IL-4-treated mice also showed a shorter duration of the first EAE attack, a longer inter-relapse period, and a reduction in the severity and duration of the first relapse. Protection from EAE progression in IL-4-treated mice was associated with activation of microglia in spinal cord areas where mRNA content of the pro-inflammatory chemokines, macrophage chemoattractant protein-1 (MCP-1) and Rantes, was reduced and that of the anti-inflammatory cytokine IL-4 was increased. Finally, CNS-infiltrating mononuclear cells from IL-4-treated mice produced lower levels of MCP-1 mRNA compared with control mice. Our results, showing that IL-4 gene delivery using HSV-1 vectors induces protection from EAE by in situ modulating the cytokine/chemokine-mediated circuits sustaining effector cell functions, indicate that the intrathecal 'therapeutic' use of nonreplicative HSV-1-derived vectors containing anti-inflammatory molecules might represent an alternative strategy in inflammatory diseases of the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Interleukin-4/genetics , Animals , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Cisterna Magna , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Genetic Vectors , Herpesvirus 1, Human/genetics , Injections, Intraventricular , Interleukin-4/biosynthesis , Mice , Mice, Inbred Strains , Microglia/pathology , Multiple Sclerosis/therapy , RNA, Messenger/genetics , Spinal Cord/metabolism
13.
J Neuroimmunol ; 111(1-2): 86-92, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11063825

ABSTRACT

Systemic administration of interferon (IFN)-beta has been recently approved for the treatment of relapsing-remitting multiple sclerosis (RRMS). The immunological mechanism by which IFN-beta ameliorates MS is still partially unknown. We measured the number of blood circulating CD4(+), CD4(-), CD8(+), and CD8(-) T cells secreting IFN-gamma and IL-4 in 26 RRMS patients followed for up to 9 months of an alternate day s.c. treatment with 8x16 IU of IFN-beta1b. Compared to pre-treatment values, a significant (P<0.05) reduction of CD4(+), CD4(-), CD8(+) and CD8(-) cells producing IFN-gamma and of CD4(+) and CD4(-) cells producing IL-4 was observed in MS patients. The IFN-beta-associated effect was evident soon after the beginning of the treatment and persisted for the entire follow-up period. We did not observe any effect of IFN-beta treatment on the percentage of IL-4-producing CD8(+) and CD8(-) cells nor in that of natural killer (NK) cells producing IFN-gamma. Our results show that IFN-beta treatment in MS patients induces a profound and persistent down-regulation of the number of circulating T cells secreting IFN-gamma and IL-4 thus suggesting a broader rather than a specific immunomodulatory effect of IFN-beta in MS.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interferon-beta/administration & dosage , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , T-Lymphocytes, Helper-Inducer/drug effects , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, IgG/analysis , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
14.
J Neuroimmunol ; 109(1): 3-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969174

ABSTRACT

Multiple sclerosis (MS) is considered an immune-mediated disease of the central nervous system (CNS) sustained by a chronic inflammatory process leading to patchy demyelination and axonal loss. However, the inflammatory triggering event as well as the target of the pathogenic process in MS are still partially unknown. We report evidence that a 'local' inflammatory process occurring in the CNS (considered as a reaction of blood vessels in vascularized living tissue to a local injury leading to the accumulation of fluid and blood cells) along with a concomitant, but possibly unrelated, peripheral inflammatory event may trigger a CNS-specific autoimmune reaction cascade sustaining the MS pathogenesis. In the CNS, inflammatory mediators (mainly cytokines) act either as regulatory (i.e. activation of glial cells, shaping the autoimmune response) or effector molecules (i.e. myelinotoxicity, oligodendrotoxicity). In the periphery, inflammatory cytokines induce, in a bystander fashion, activation of monocytes and T cells. Among this latter cell population there are myelin-specific T cells belonging to the normal 'autoimmune' repertoire that home to the CNS where they may trigger the continuous recruitment of effector cells (macrophages) from the periphery. The concept that two concomitant, but possibly unrelated, inflammatory events, occurring in the CNS and in the periphery, represent the crucial elements sustaining MS, might reveal a more comprehensive view (dual signal hypothesis) of the entire etiopathogenic process underlying this disease.


Subject(s)
Cytokines/physiology , Multiple Sclerosis/immunology , Animals , Brain/immunology , Humans , Inflammation/etiology , Inflammation Mediators/physiology , Lymphocyte Activation , Multiple Sclerosis/etiology , T-Lymphocytes/immunology
15.
Neurol Sci ; 21(4 Suppl 2): S871-5, 2000.
Article in English | MEDLINE | ID: mdl-11205366

ABSTRACT

Multiple sclerosis (MS) is characterized by the presence in the central nervous system (CNS) of perivascular inflammatory infiltrates containing, among others, autoreactive T cells and activated macrophages. These observations indicate that MS is a T cell-mediated CNS-confined chronic inflammatory demyelinating disease in which the ultimate effector cell is the activated macrophage. The inflammatory process, leading to patchy demyelination and axonal loss, is mainly sustained by pro-inflammatory cytokines that, along with chemokines, adhesion molecules and metalloproteases, modulate at different levels the pathogenic process underlying MS. Due to their central role in MS pathogenesis, "inflammatory" molecules might represent suitable peripheral markers of disease (disease-trait) and/or disease activity (state-trait). However, reliable disease-trait or state-trait immunological markers for MS have not yet been identified. The intrinsic characteristics of these molecules (i.e. autocrine/paracrine activity, short half-life, redundancy) may in part explain their inconsistency as disease markers. Additionally, the unreliability of methodologies and the lack of careful patient stratification can also, at least in part, account for the unsatisfactory results so far obtained.


Subject(s)
Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Biomarkers/blood , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/immunology , Chemokines/blood , Chemokines/immunology , Cysteine Endopeptidases/blood , Cysteine Endopeptidases/immunology , Cytokines/blood , Cytokines/immunology , Humans , Matrix Metalloproteinases/blood , Matrix Metalloproteinases/immunology
16.
Magn Reson Imaging ; 17(10): 1521-3, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10610001

ABSTRACT

A method for intracellular iron labeling of human mononuclear cells (lymphocytes and monocytes) for magnetic resonance imaging (MRI) using simple incubation of cells with approved MRI iron contrast agents is presented. Labeled cells can be detected by MRI in vitro, and this suggests the possibility that the technique could become a marker for in vivo lymphocyte and monocyte trafficking studies in acute inflammatory lesions such as those in Multiple Sclerosis.


Subject(s)
Contrast Media , Iron , Lymphocytes/pathology , Magnetic Resonance Imaging/methods , Monocytes/pathology , Multiple Sclerosis/diagnosis , Oxides , Cell Survival , Cells, Cultured , Dextrans , Ferrosoferric Oxide , Humans , Intracellular Fluid , Magnetite Nanoparticles , Reproducibility of Results , Suspensions
17.
J Neurol Neurosurg Psychiatry ; 67(6): 785-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567499

ABSTRACT

The cysteine protease caspase-1 plays a crucial part in the inflammatory process due to its ability to proteolitically activate proinflammatory cytokine precursors, such as interleukin (IL)-1beta and IL-18. Multiple sclerosis is a chronic inflammatory demyelinating disease of the CNS in which the pathogenic process is mainly orchestrated by proinflammatory cytokines. The role of caspase-1 in multiple sclerosis was evaluated by measuring its mRNA levels in peripheral blood mononuclear cells (PBMCs) from seven patients with relapsing-remitting multiple sclerosis every 15 days over a 1 year period. The recorded levels were compared with clinical and MRI evidence of disease activity. Brain MRI was performed monthly in each patient. Caspase-1 mRNA levels were significantly increased in PBMCs from patients with multiple sclerosis compared with healthy controls (p<0.001). In patients with multiple sclerosis, a twofold to threefold increase of caspase-1 mRNA mean level was found in the week preceding an acute attack (p<0. 05). The magnitude of caspase-1 mRNA increase correlated with the number of new (p=0.01) but not persisting gadolinium enhancing brain MRI lesions. In conclusion, caspase-1 might be involved in the immune mediated process underlying CNS inflammation and might represent a suitable peripheral immunological marker of disease activity in multiple sclerosis.


Subject(s)
Brain/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Leukocytes, Mononuclear/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , RNA, Messenger/genetics , Adult , Brain/pathology , DNA, Complementary/genetics , Female , Genetic Markers , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Magnetic Resonance Imaging , Male , Protein Precursors/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Severity of Illness Index
18.
J Immunol ; 163(5): 2403-9, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10452974

ABSTRACT

T cell-mediated inflammation is considered to play a key role in the pathogenic mechanisms sustaining multiple sclerosis (MS). Caspase-1, formerly designated IL-1beta-converting enzyme, is crucially involved in immune-mediated inflammation because of its pivotal role in regulating the cellular export of IL-1beta and IL-18. We studied the role of caspase-1 in experimental autoimmune encephalomyelitis (EAE), the animal model for MS. Caspase-1 is transcriptionally induced during EAE, and its levels correlate with the clinical course and transcription rate of proinflammatory cytokines such as TNF-alpha, IL-1beta, IFN-gamma, and IL-6. A reduction of EAE incidence and severity is observed in caspase-1-deficient mice, depending on the immunogenicity and on the amount of the encephalitogenic myelin oligodendrocyte glycoprotein (MOG) peptide used. In caspase-1-deficient mice, reduced EAE incidence correlates with defective development of anti-MOG IFN-gamma-producing Th1 cells. Finally, pharmacological blockade of caspase-1 in Biozzi AB/H mice, immunized with spinal cord homogenate or MOG35-55 peptide, by the caspase-1-inhibitor Z-Val-Ala-dl -Asp-fluoromethylketone, significantly reduces EAE incidence in a preventive but not in a therapeutic protocol. These results indicate that caspase-1 plays an important role in the early stage of the immune-mediated inflammatory process leading to EAE, thus representing a possible therapeutic target in the acute phase of relapsing remitting MS.


Subject(s)
Caspase 1/physiology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Sheath/physiology , Spinal Cord/pathology , Spinal Cord/physiopathology , Amino Acid Chloromethyl Ketones/administration & dosage , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/physiopathology , Caspase 1/deficiency , Caspase 1/genetics , Caspase Inhibitors , Cysteine Proteinase Inhibitors/administration & dosage , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Immunosuppressive Agents/administration & dosage , Infusion Pumps, Implantable , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/pathology , Neuroprotective Agents/administration & dosage , RNA, Messenger/biosynthesis , Spinal Cord/immunology , Th1 Cells/enzymology , Th1 Cells/immunology , Up-Regulation/immunology
19.
Clin Chem ; 45(3): 400-5, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10053042

ABSTRACT

BACKGROUND: Acetylcholine receptor (AChR) from human muscles is the antigen used currently in radioimmunoprecipitation assays (RIPAs) for the determination of anti-AChR antibodies in the diagnosis of myasthenia gravis (MG). Our aim was to develop and validate an ELISA using TE671 cells as the source of AChR. METHODS: After TE671 cell homogenization, the crude AChR extract was used for plate coating. Anti-AChR antibodies were determined in 207 MG patients and in 77 controls. RESULTS: The mean intra- and interassay CVs (for two samples with different anti-AChR antibody concentrations) were 9.7% and 15.7%, respectively. Test sensitivity and specificity, for generalized MG, were 79.5% (95% confidence interval, 72.8-85.0%) and 96.1% (89.0-99.1%). The detection limit was 2 nmol/L. Anti-AChR antibody concentrations from 53 MG patients, as tested with our ELISA, showed good agreement with an RIPA with a mean difference (SD) of 1.0 (5.6) nmol/L. CONCLUSION: Our ELISA is a simple screening test for the diagnosis of MG and enables rapid and inexpensive patient follow-up.


Subject(s)
Antibodies/blood , Myasthenia Gravis/immunology , Receptors, Cholinergic/immunology , Adolescent , Adult , Calibration , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male , Middle Aged , Myasthenia Gravis/blood , Reproducibility of Results , Sensitivity and Specificity , Tumor Cells, Cultured
20.
Hum Gene Ther ; 9(17): 2605-17, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9853527

ABSTRACT

Multiple sclerosis (MS) is a T cell-mediated organ-specific inflammatory disease leading to central nervous system (CNS) demyelination. On the basis of results obtained in experimental autoimmune encephalomyelitis (EAE) models, MS treatment by administration of antiinflammatory cytokines such as interleukin 4 (IL-4) is promising but is hampered by the limited access of the cytokines to the CNS and by the pleiotropic effects of systemically administered cytokines. We established a cytokine delivery system within the CNS using non-replicative herpes simplex type 1 (HSV-1) viral vectors engineered with cytokine genes. These vectors injected into the cisterna magna (i.c.) of mice diffuse in all ventricular and subarachnoid spaces and infect with high efficiency the ependymal and leptomeningeal cell layers surrounding these areas, without obvious toxic effects. Heterologous genes contained in the vectors are efficiently transcribed in infected ependymal cells, leading to the production of high amounts of the coded proteins. For example, 4.5 ng of interferon gamma (IFN-gamma) per milliliter is secreted into the cerebrospinal fluid (CSF) up to day 28 postinjection (p.i.) and reaches the CNS parenchyma in bioactive form, as demonstrated by upregulation of MHC class I expression on CNS-resident cells. We then exploited the therapeutic potential of the vectors in EAE mice. An HSV-1-derived vector containing the IL-4 gene was injected i.c. in Biozzi AB/H mice at the time of EAE induction. We found the following in treated mice: (1) delayed EAE onset, (2) a significant decrease in clinical score, (3) a significant decrease in perivascular inflammatory infiltrates and in the number of macrophages infiltrating the CNS parenchyma and the submeningeal spaces, and (4) a reduction in demyelinated areas and axonal loss. Peripheral T cells from IL-4-treated mice were not affected either in their antigen-specific proliferative response or in cytokine secretion pattern. Our results indicate that CNS cytokine delivery with HSV-1 vectors is feasible and might represent an approach for the treatment of demyelinating diseases. Advantages of this approach over systemic cytokine administration are the high cytokine level reached in the CNS, the absence of effects on the peripheral immune system, and the long-lasting cytokine production in the CNS after a single vector administration.


Subject(s)
Central Nervous System/metabolism , Defective Viruses/genetics , Demyelinating Diseases/prevention & control , Encephalomyelitis, Autoimmune, Experimental/pathology , Herpesvirus 1, Human/genetics , Interleukin-4/administration & dosage , Animals , Cell Division , Central Nervous System/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Vectors , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...