Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Ann Neurol ; 90(5): 789-807, 2021 11.
Article in English | MEDLINE | ID: mdl-34476836

ABSTRACT

OBJECTIVE: Parkinson's disease (PD) manifests in motor dysfunction, non-motor symptoms, and eventual dementia (PDD). Neuropathological hallmarks include nigrostriatal neurodegeneration, Lewy body (LB) pathology, and neuroinflammation. Alpha-synuclein (α-syn), a primary component of LBs, is implicated in PD pathogenesis, accumulating, and aggregating in both familial and sporadic PD. However, as α-syn pathology is often comorbid with amyloid-beta (Aß) plaques and phosphorylated tau (pTau) tangles in PDD, it is still unclear whether α-syn is the primary cause of neurodegeneration in sporadic PDD. We aimed to determine how the absence of α-syn would affect PDD manifestation. METHODS: IFN-ß knockout (Ifnb-/- ) mice spontaneously develop progressive behavior abnormalities and neuropathology resembling PDD, notably with α-syn+ LBs. We generated Ifnb/Snca double knockout (DKO) mice and evaluated their behavior and neuropathology compared with wild-type (Wt), Ifnb-/- , and Snca-/- mice using immunohistochemistry, electron microscopy, immunoblots, qPCR, and modification of neuronal signaling. RESULTS: Ifnb/Snca DKO mice developed all clinical PDD-like behavioral manifestations induced by IFN-ß loss. Independently of α-syn expression, lack of IFN-ß alone induced Aß plaques, pTau tangles, and LB-like Aß+ /pTau+ inclusion bodies and neuroinflammation. IFN-ß loss caused significant elevated glial and neuronal TNF-α and neuronal TNFR1, associated with neurodegeneration. Restoring neuronal IFN-ß signaling or blocking TNFR1 rescued caspase 3/t-BID-mediated neuronal-death through upregulation of c-FLIPS and lowered intraneuronal Aß and pTau accumulation. INTERPRETATION: These findings increase our understanding of PD pathology and suggest that targeting α-syn alone is not sufficient to mitigate disease. Targeting specific aspects of neuroinflammation, such as aberrant neuronal TNF-α/TNFR1 or IFN-ß/IFNAR signaling, may attenuate disease. ANN NEUROL 2021;90:789-807.


Subject(s)
Neuroinflammatory Diseases/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Progression , Lewy Bodies/pathology , Lewy Body Disease/metabolism , Mice, Knockout , Neuroglia/pathology , Parkinson Disease/genetics , Plaque, Amyloid/metabolism , Tumor Necrosis Factor-alpha/deficiency
2.
Sci Transl Med ; 10(461)2018 10 03.
Article in English | MEDLINE | ID: mdl-30282695

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutation in the huntingtin (HTT) protein, resulting in acquisition of toxic functions. Previous studies have shown that lowering mutant HTT has the potential to be broadly beneficial. We previously identified HTT single-nucleotide polymorphisms (SNPs) tightly linked to the HD mutation and developed antisense oligonucleotides (ASOs) targeting HD-SNPs that selectively suppress mutant HTT. We tested allele-specific ASOs in a mouse model of HD. Both early and late treatment reduced cognitive and behavioral impairments in mice. To determine the translational potential of the treatment, we examined the effect of ASO administration on HTT brain expression in nonhuman primates. The treatment induced robust HTT suppression throughout the cortex and limbic system, areas implicated in cognition and psychiatric function. The results suggest that ASOs specifically targeting mutated HTT might have therapeutic effects on HD-mediated cognitive impairments.


Subject(s)
Cognition , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/physiopathology , Animals , Anxiety/complications , Anxiety/pathology , Anxiety/physiopathology , Atrophy/pathology , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Female , Humans , Huntington Disease/complications , Huntington Disease/pathology , Limbic System/pathology , Male , Mutant Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Primates
3.
Hum Mol Genet ; 27(2): 239-253, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29121340

ABSTRACT

Oxidative stress is a prominent feature of Huntington disease (HD), and we have shown previously that reduced levels of hace1 (HECT domain and Ankyrin repeat containing E3 ubiquitin protein ligase 1) in patient striatum may contribute to the pathogenesis of HD. Hace1 promotes the stability of Nrf2 and thus plays an important role in antioxidant response mechanisms, which are dysfunctional in HD. Moreover, hace1 overexpression mitigates mutant huntingtin (mHTT)-induced oxidative stress in vitro through promotion of the Nrf2 antioxidant response. Here, we show that the genetic ablation of hace1 in the YAC128 mouse model of HD accelerates motor deficits and exacerbates cognitive and psychiatric phenotypes in vivo. We find that both the expression of mHTT and the ablation of hace1 alone are sufficient to cause deficits in astrocytic mitochondrial respiration. We confirm the crucial role of hace1 in astrocytes in vivo, since its ablation is sufficient to cause dramatic astrogliosis in wild-type FVB/N mice. Astrogliosis is not observed in the presence of mHTT but a strong dysregulation in the expression of astrocytic markers in HACE1-/- x YAC128 striatum suggests an additive effect of mHTT expression and hace1 loss on this cell type. HACE1-/- x YAC128 mice and primary cells derived from these animals therefore provide model systems that will allow for the further dissection of Nrf2 pathways and astrocyte dysfunction in the context of HD.


Subject(s)
Astrocytes/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Neostriatum/metabolism , Nerve Tissue Proteins/metabolism , Oxidative Stress/physiology
4.
Hum Mol Genet ; 26(6): 1115-1132, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28104789

ABSTRACT

Huntington disease (HD) is a neurodegenerative disease caused by a mutation in the huntingtin (HTT) gene. HTT is a large protein, interacts with many partners and is involved in many cellular pathways, which are perturbed in HD. Therapies targeting HTT directly are likely to provide the most global benefit. Thus there is a need for preclinical models of HD recapitulating human HTT genetics. We previously generated a humanized mouse model of HD, Hu97/18, by intercrossing BACHD and YAC18 mice with knockout of the endogenous mouse HD homolog (Hdh). Hu97/18 mice recapitulate the genetics of HD, having two full-length, genomic human HTT transgenes heterozygous for the HD mutation and polymorphisms associated with HD in populations of Caucasian descent. We have now generated a companion model, Hu128/21, by intercrossing YAC128 and BAC21 mice on the Hdh-/- background. Hu128/21 mice have two full-length, genomic human HTT transgenes heterozygous for the HD mutation and polymorphisms associated with HD in populations of East Asian descent and in a minority of patients from other ethnic groups. Hu128/21 mice display a wide variety of HD-like phenotypes that are similar to YAC128 mice. Additionally, both transgenes in Hu128/21 mice match the human HTT exon 1 reference sequence. Conversely, the BACHD transgene carries a floxed, synthetic exon 1 sequence. Hu128/21 mice will be useful for investigations of human HTT that cannot be addressed in Hu97/18 mice, for developing therapies targeted to exon 1, and for preclinical screening of personalized HTT lowering therapies in HD patients of East Asian descent.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Mutation/genetics , Alleles , Animals , Disease Models, Animal , Exons/genetics , Heterozygote , Humans , Huntington Disease/pathology , Mice , Mice, Transgenic , Phenotype
5.
Hum Mol Genet ; 25(17): 3654-3675, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27378694

ABSTRACT

Huntington disease (HD) model mice with heterozygous knock-in (KI) of an expanded CAG tract in exon 1 of the mouse huntingtin (Htt) gene homolog genetically recapitulate the mutation that causes HD, and might be favoured for preclinical studies. However, historically these mice have failed to phenotypically recapitulate the human disease. Thus, homozygous KI mice, which lack wildtype Htt, and are much less relevant to human HD, have been used. The zQ175 model was the first KI mouse to exhibit significant HD-like phenotypes when heterozygous. In an effort to exacerbate HD-like phenotypes and enhance preclinical utility, we have backcrossed zQ175 mice to FVB/N, a strain highly susceptible to neurodegeneration. These Q175F mice display significant HD-like phenotypes along with sudden early death from fatal seizures. The zQ175 KI allele retains a floxed neomycin resistance cassette upstream of the Htt gene locus and produces dramatically reduced mutant Htt as compared to the endogenous wildtype Htt allele. By intercrossing with mice expressing cre in germ line cells, we have excised the neo cassette from Q175F mice generating a new line, Q175FΔneo (Q175FDN). Removal of the neo cassette resulted in a ∼2 fold increase in mutant Htt and rescue of fatal seizures, indicating that the early death phenotype of Q175F mice is caused by Htt deficiency rather than by mutant Htt. Additionally, Q175FDN mice exhibit earlier onset and a greater variety and severity of HD-like phenotypes than Q175F mice or any previously reported KI HD mouse model, making them valuable for preclinical studies.


Subject(s)
Gene Knock-In Techniques/methods , Huntingtin Protein/genetics , Huntington Disease/genetics , Mutation , Animals , Behavior, Animal , Crosses, Genetic , Disease Models, Animal , Heterozygote , Humans , Huntington Disease/pathology , Mice , Phenotype
6.
Sci Rep ; 5: 12166, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26174131

ABSTRACT

Quantitation of huntingtin protein in the brain is needed, both as a marker of Huntington disease (HD) progression and for use in clinical gene silencing trials. Measurement of huntingtin in cerebrospinal fluid could be a biomarker of brain huntingtin, but traditional protein quantitation methods have failed to detect huntingtin in cerebrospinal fluid. Using micro-bead based immunoprecipitation and flow cytometry (IP-FCM), we have developed a highly sensitive mutant huntingtin detection assay. The sensitivity of huntingtin IP-FCM enables accurate detection of mutant huntingtin protein in the cerebrospinal fluid of HD patients and model mice, demonstrating that mutant huntingtin levels in cerebrospinal fluid reflect brain levels, increasing with disease stage and decreasing following brain huntingtin suppression. This technique has potential applications as a research tool and as a clinical biomarker.


Subject(s)
Brain/metabolism , Huntington Disease/pathology , Nerve Tissue Proteins/cerebrospinal fluid , Adult , Aged , Animals , Brain/pathology , Disease Models, Animal , Female , Flow Cytometry , Humans , Huntingtin Protein , Huntington Disease/metabolism , Immunoblotting , Immunoprecipitation , Male , Mice , Middle Aged , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Severity of Illness Index
7.
Neurobiol Dis ; 76: 46-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25662335

ABSTRACT

Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Huntington Disease/therapy , Semaphorins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Brain/metabolism , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/therapy , Disease Models, Animal , Huntington Disease/complications , Immunotherapy , Mice , Mice, Transgenic , Motor Activity/drug effects , Signal Transduction/drug effects
8.
Mol Ther ; 22(12): 2093-2106, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25101598

ABSTRACT

Huntington disease (HD) is a dominant, genetic neurodegenerative disease characterized by progressive loss of voluntary motor control, psychiatric disturbance, and cognitive decline, for which there is currently no disease-modifying therapy. HD is caused by the expansion of a CAG tract in the huntingtin (HTT) gene. The mutant HTT protein (muHTT) acquires toxic functions, and there is significant evidence that muHTT lowering would be therapeutically efficacious. However, the wild-type HTT protein (wtHTT) serves vital functions, making allele-specific muHTT lowering strategies potentially safer than nonselective strategies. CAG tract expansion is associated with single nucleotide polymorphisms (SNPs) that can be targeted by gene silencing reagents such as antisense oligonucleotides (ASOs) to accomplish allele-specific muHTT lowering. Here we evaluate ASOs targeted to HD-associated SNPs in acute in vivo studies including screening, distribution, duration of action and dosing, using a humanized mouse model of HD, Hu97/18, that is heterozygous for the targeted SNPs. We have identified four well-tolerated lead ASOs that potently and selectively silence muHTT at a broad range of doses throughout the central nervous system for 16 weeks or more after a single intracerebroventricular (ICV) injection. With further validation, these ASOs could provide a therapeutic option for individuals afflicted with HD.


Subject(s)
Brain/pathology , Huntington Disease/therapy , Mutant Proteins/metabolism , Nerve Tissue Proteins/genetics , Oligonucleotides, Antisense/administration & dosage , Thionucleotides/administration & dosage , Animals , Brain/metabolism , Disease Models, Animal , Gene Silencing , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Injections , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Nerve Tissue Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Polymorphism, Single Nucleotide , Rats , Rats, Sprague-Dawley , Thionucleotides/pharmacology
9.
Mol Cell Neurosci ; 60: 88-96, 2014 May.
Article in English | MEDLINE | ID: mdl-24769106

ABSTRACT

Microglia represent mononuclear phagocytes in the brain and perform immune surveillance, recognizing a number of signaling molecules released from surrounding cells in both healthy and pathological situations. The microglia interact with several damage-associated molecular pattern molecules (DAMPs) and recent data indicate that mitochondrial transcription factor A (Tfam) could act as a specific DAMP in peripheral tissues. This study tested the hypothesis that extracellular Tfam induces pro-inflammatory and cytotoxic responses of the microglia. Three different types of human mononuclear phagocytes were used to model human microglia: human peripheral blood monocytes from healthy donors, human THP-1 monocytic cells, and human primary microglia obtained from autopsy samples. When combined with interferon (IFN)-γ, recombinant human Tfam (rhTfam) induced secretions that were toxic to human SH-SY5Y neuroblastoma cells in all three models. Similar cytotoxic responses were observed when THP-1 cells and human microglia were exposed to human mitochondrial proteins in the presence of IFN-γ. rhTfam alone induced expression of pro-inflammatory cytokines interleukin (IL)-1ß, IL-6 and IL-8 by THP-1 cells. This induction was further enhanced in the presence of IFN-γ. Upregulated secretion of IL-6 in response to rhTfam plus IFN-γ was confirmed in primary human microglia. Use of specific inhibitors showed that the rhTfam-induced cytotoxicity of human THP-1 cells depended partially on activation of c-Jun N-terminal kinase (JNK), but not p38 mitogen-activated protein kinase (MAPK). Overall, our data support the hypothesis that, in the human brain, Tfam could act as an intercellular signaling molecule that is recognized by the microglia to cause pro-inflammatory and cytotoxic responses.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/metabolism , MAP Kinase Signaling System , Microglia/metabolism , Mitochondrial Proteins/metabolism , Transcription Factors/metabolism , Brain/cytology , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/toxicity , Extracellular Space/metabolism , Humans , Inflammation/metabolism , Interferon-gamma/pharmacology , Interleukins/genetics , Interleukins/metabolism , Microglia/drug effects , Mitochondrial Proteins/toxicity , Monocytes/metabolism , Transcription Factors/toxicity
10.
Nucleic Acids Res ; 41(21): 9634-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23963702

ABSTRACT

Autosomal dominant diseases such as Huntington's disease (HD) are caused by a gain of function mutant protein and/or RNA. An ideal treatment for these diseases is to selectively suppress expression of the mutant allele while preserving expression of the wild-type variant. RNase H active antisense oligonucleotides (ASOs) or small interfering RNAs can achieve allele selective suppression of gene expression by targeting single nucleotide polymorphisms (SNPs) associated with the repeat expansion. ASOs have been previously shown to discriminate single nucleotide changes in targeted RNAs with ∼5-fold selectivity. Based on RNase H enzymology, we enhanced single nucleotide discrimination by positional incorporation of chemical modifications within the oligonucleotide to limit RNase H cleavage of the non-targeted transcript. The resulting oligonucleotides demonstrate >100-fold discrimination for a single nucleotide change at an SNP site in the disease causing huntingtin mRNA, in patient cells and in a completely humanized mouse model of HD. The modified ASOs were also well tolerated after injection into the central nervous system of wild-type animals, suggesting that their tolerability profile is suitable for advancement as potential allele-selective HD therapeutics. Our findings lay the foundation for efficient allele-selective downregulation of gene expression using ASOs-an outcome with broad application to HD and other dominant genetic disorders.


Subject(s)
Alleles , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Oligonucleotides, Antisense/chemistry , Polymorphism, Single Nucleotide , Animals , Base Pairing , Brain/metabolism , Cells, Cultured , Down-Regulation , Fluorine/chemistry , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Oligonucleotides, Antisense/administration & dosage , Rats , Rats, Sprague-Dawley , Ribonuclease H/metabolism
11.
Hum Mol Genet ; 22(1): 18-34, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23001568

ABSTRACT

Silencing the mutant huntingtin gene (muHTT) is a direct and simple therapeutic strategy for the treatment of Huntington disease (HD) in principle. However, targeting the HD mutation presents challenges because it is an expansion of a common genetic element (a CAG tract) that is found throughout the genome. Moreover, the HTT protein is important for neuronal health throughout life, and silencing strategies that also reduce the wild-type HTT allele may not be well tolerated during the long-term treatment of HD. Several HTT silencing strategies are in development that target genetic sites in HTT that are outside of the CAG expansion, including HD mutation-linked single-nucleotide polymorphisms and the HTT promoter. Preclinical testing of these genetic therapies has required the development of a new mouse model of HD that carries these human-specific genetic targets. To generate a fully humanized mouse model of HD, we have cross-bred BACHD and YAC18 on the Hdh(-/-) background. The resulting line, Hu97/18, is the first murine model of HD that fully genetically recapitulates human HD having two human HTT genes, no mouse Hdh genes and heterozygosity of the HD mutation. We find that Hu97/18 mice display many of the behavioral changes associated with HD including motor, psychiatric and cognitive deficits, as well as canonical neuropathological abnormalities. This mouse line will be useful for gaining additional insights into the disease mechanisms of HD as well as for testing genetic therapies targeting human HTT.


Subject(s)
Disease Models, Animal , Huntington Disease/genetics , Animals , Gene Silencing , Humans , Huntington Disease/psychology , Mice , Mice, Transgenic , Mutation , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Rotarod Performance Test
12.
Mol Cell Neurosci ; 49(4): 430-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22406427

ABSTRACT

Neuroinflammation, which is one of the hallmarks of neurodegenerative disorders such as Alzheimer's disease, involves secretion of pro-inflammatory mediators by activated glial cells. Secreted phospholipase A(2) group IIA (sPLA(2)IIA) has been implicated as an inflammatory mediator contributing to various peripheral inflammatory conditions; however, little is known about the role this enzyme plays in neuroinflammation. Human microglia-like promonocytic THP-1 cells and human primary astrocytes were used to study sPLA(2)IIA expression, secretion and function. Production of sPLA(2)IIA by these cells was induced in response to stimulation by pro-inflammatory mediators at both mRNA and protein levels. Removal of sPLA(2)IIA from stimulated human microglia-like cell and astrocyte supernatants by immunosorbent caused significant reduction of their toxicity towards SH-SY5Y neuroblastoma cells. Both sPLA(2)IIA specific and non-specific PLA(2) inhibitors exhibited no anti-cytotoxic or neuroprotective effects, suggesting that sPLA(2)IIA cytotoxicity is mediated by a non-enzymatic mechanism. The data obtained indicate that sPLA(2)IIA may contribute to the pathogenesis of neurodegenerative diseases involving neuroinflammation. Agents inhibiting the non-enzymatic actions of sPLA(2)IIA could be used to slow down progression of neurodegenerative processes that are driven by inflammation.


Subject(s)
Group II Phospholipases A2/metabolism , Neuroglia/metabolism , Neurons/drug effects , Neurotoxins/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Group II Phospholipases A2/pharmacology , Humans , Inflammation/metabolism , Neuroglia/drug effects , Neurons/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
13.
Brain Res Bull ; 84(3): 244-51, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21185917

ABSTRACT

In vitro cultures of various glial cell types are common systems used to model neuroinflammatory processes associated with age-dependent human neurodegenerative diseases. Even though most researchers choose to use neonatal rodent brain tissues as the source of glial cells, there are significant variations in glial cell functions that are species and age dependent. It has been established that human and swine immune systems have a number of similarities, which suggests that cultured porcine microglia and astrocytes may be good surrogates for human glial cell types. Here we describe a method that could be used to prepare more than 90% pure microglia and astrocyte cultures derived from adult porcine tissues. We demonstrate that both microglia and astrocytes derived from adult porcine brains express functional interferon-γ receptors (IFN-γ-R) and CD14. They become toxic towards SH-SY5Y neuroblastoma cells when exposed to proinflammatory mediators. Upon such stimulation with lipopolysaccharide (LPS) and interferon-γ (IFN-γ), adult porcine microglia, but not astrocytes, secrete tumor necrosis factor-α (TNF-α) while both cell types do not secrete detectable levels of nitric oxide (NO). Comparison of our experimental data with previously published studies indicates that adult porcine glial cultures have certain functional characteristics that make them similar to human glial cells. Therefore adult porcine glial cells may be a useful model system for studies of human diseases associated with adulthood and advanced age. Adult porcine tissues are relatively easy to obtain in most countries and could be used as a reliable and inexpensive source of cultured cells.


Subject(s)
Astrocytes/physiology , Microglia/physiology , Receptors, Interferon/physiology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Lineage/immunology , Cells, Cultured , Humans , Microglia/immunology , Microglia/metabolism , Receptors, Interferon/biosynthesis , Sus scrofa , Interferon gamma Receptor
14.
J Neuroimmunol ; 233(1-2): 65-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21185608

ABSTRACT

The effect of a moderate, physiologically relevant rise in temperature on several neuroinflammatory parameters was investigated in vitro using human cell lines and cultured human astrocytes. A two degree Celsius rise in temperature was found to enhance the neurotoxicity of microglia-like and astrocytic cells, increase the release of monocyte chemotactic protein (MCP)-1 by activated human monocytic THP-1 cells and amplify the generation of reactive oxygen intermediates by differentiated HL-60 myelocytic cells. Moderate increases in body temperature may exacerbate neuroinflammation and neuronal injury in chronic neurodegenerative disorders. Hence, therapies aimed at lowering the body temperature could be used to slow down the progression of such diseases.


Subject(s)
Body Temperature/immunology , Brain/immunology , Brain/pathology , Fever/immunology , Neuroimmunomodulation/immunology , Neurons/immunology , Neurons/pathology , Cell Culture Techniques/methods , Cells, Cultured , Fever/pathology , HL-60 Cells , Humans
15.
Bioorg Med Chem ; 18(15): 5685-96, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20609589

ABSTRACT

A novel dipyrazole ethandiamide compound and acid chloride of pyrazolo[3,4-d]pyrimidine 4(5H)-one were prepared and reacted with a number of nucleophiles. The resultant novel compounds were tested in several in vitro and in vivo assays. Three compounds inhibited the secretion of neurotoxins by human THP-1 monocytic cells at concentrations that were not toxic to these cells. They also partially inhibited both cyclooxygenase-1 and -2 isoforms. In animal studies, two compounds were notable for their anti-inflammatory activity that was comparable to that of the clinically available cyclooxygenase-2 inhibitor celecoxib. Modeling studies by using the molecular operating environment module showed comparable docking scores for the two enantiomers docked in the active site of cyclooxygenase-2.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Pyrazoles/chemistry , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Binding Sites , Catalytic Domain , Celecoxib , Cell Line , Computer Simulation , Crystallography, X-Ray , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/toxicity , Humans , Models, Molecular , Pyrazoles/chemical synthesis , Pyrazoles/toxicity , Sulfonamides/chemistry , Sulfonamides/toxicity
16.
Bioorg Med Chem ; 18(5): 2019-28, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20138770

ABSTRACT

Novel pyrazolyl-2,4-thiazolidinediones were prepared via the reaction of appropriate pyrazolecarboxaldehydes with 2,4-thiazolidinediones and substituted benzyl-2,4-thiazolidinediones. The resultant compounds were first evaluated for their anti-inflammatory and neuroprotective properties in vitro. The active compounds were further studied in vivo by using the formalin-induced paw edema and the turpentine oil-induced granuloma pouch bioassays. We identified four novel compounds that showed protective effects in vitro at non-toxic concentrations, and were also effective in the animal models of acute and sub-acute inflammation.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Neuroprotective Agents/chemical synthesis , Thiazolidinediones/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Cell Line, Tumor , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Edema/chemically induced , Edema/drug therapy , Granuloma/chemically induced , Granuloma/drug therapy , HL-60 Cells , Humans , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Rats , Thiazolidinediones/chemical synthesis , Thiazolidinediones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...