Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cytokine ; 102: 83-93, 2018 02.
Article in English | MEDLINE | ID: mdl-28800924

ABSTRACT

Macrophages are common targets for infection and innate immune activation by many pathogenic viruses including the neurotropic Theiler's Murine Encephalomyelitis Virus (TMEV). As both infection and innate activation of macrophages are key determinants of viral pathogenesis especially in the central nervous system (CNS), an analysis of macrophage growth factors on these events was performed. C3H mouse bone-marrow cells were differentiated in culture using either recombinant macrophage colony stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF), inoculated with TMEV (BeAn) and analyzed at various times thereafter. Cytokine RNA and protein analysis, virus titers, and flow cytometry were performed to characterize virological parameters under these culture conditions. GM-CSF-differentiated macrophages showed higher levels of TMEV viral RNA and proinflammatory molecules compared to infected M-CSF-differentiated cells. Thus, GM-CSF increases both TMEV infection and TMEV-induced activation of macrophages compared to that seen with M-CSF. Moreover, while infectious viral particles decreased from a peak at 12h to undetectable levels at 48h post infection, TMEV viral RNA remained higher in GM-CSF- compared to M-CSF-differentiated macrophages in concert with increased proinflammatory gene expression. Analysis of a possible basis for these differences determined that glycolytic rates contributed to heightened virus replication and proinflammatory cytokine secretion in GM-CSF compared to M-CSF-differentiated macrophages. In conclusion, we provide evidence implicating a role for GM-CSF in promoting virus replication and proinflammatory cytokine expression in macrophages, indicating that GM-CSF may be a key factor for TMEV infection and the induction of chronic TMEV-induced immunopathogenesis in the CNS.


Subject(s)
Cardiovirus Infections/etiology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Macrophage Activation , Macrophage Colony-Stimulating Factor/immunology , Theilovirus/pathogenicity , Animals , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Cell Differentiation/immunology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Glycolysis , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , Theilovirus/genetics , Theilovirus/isolation & purification , Virus Replication/immunology
2.
J Immunol ; 194(6): 2796-809, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681345

ABSTRACT

Virus-induced myositis is an emerging global affliction that remains poorly characterized with few treatment options. Moreover, muscle-tropic viruses often spread to the CNS, causing dramatically increased morbidity. Therefore, there is an urgent need to explore genetic factors involved in this class of human disease. This report investigates critical innate immune pathways affecting murine virus-induced myositis. Of particular importance, the key immune regulator src homology region 2 domain-containing phosphatase 1 (SHP-1), which normally suppresses macrophage-mediated inflammation, is a major factor in promoting clinical disease in muscle. We show that Theiler's murine encephalomyelitis virus (TMEV) infection of skeletal myofibers induces inflammation and subsequent dystrophic calcification, with loss of ambulation in wild-type (WT) mice. Surprisingly, although similar extensive myofiber infection and inflammation are observed in SHP-1(-/-) mice, these mice neither accumulate dead calcified myofibers nor lose ambulation. Macrophages were the predominant effector cells infiltrating WT and SHP-1(-/-) muscle, and an increased infiltration of immature monocytes/macrophages correlated with an absence of clinical disease in SHP-1(-/-) mice, whereas mature M1-like macrophages corresponded with increased myofiber degeneration in WT mice. Furthermore, blocking SHP-1 activation in WT macrophages blocked virus-induced myofiber degeneration, and pharmacologic ablation of macrophages inhibited muscle calcification in TMEV-infected WT animals. These data suggest that, following TMEV infection of muscle, SHP-1 promotes M1 differentiation of infiltrating macrophages, and these inflammatory macrophages are likely involved in damaging muscle fibers. These findings reveal a pathological role for SHP-1 in promoting inflammatory macrophage differentiation and myofiber damage in virus-infected skeletal muscle, thus identifying SHP-1 and M1 macrophages as essential mediators of virus-induced myopathy.


Subject(s)
Cardiovirus Infections/immunology , Cell Differentiation/immunology , Macrophages/immunology , Myositis/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Theilovirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cell Differentiation/genetics , Flow Cytometry , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/virology , Myositis/genetics , Myositis/virology , Oligonucleotide Array Sequence Analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Theilovirus/physiology , Transcriptome/immunology , Virus Replication/immunology
3.
Glia ; 63(10): 1753-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25919645

ABSTRACT

We have previously described reduced myelination and corresponding myelin basic protein (MBP) expression in the central nervous system of Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) deficient motheaten (me/me) mice compared with normal littermate controls. Deficiency in myelin and MBP expression in both brains and spinal cords of motheaten mice correlated with reduced MBP mRNA expression levels in vivo and in purified oligodendrocytes in vitro. Therefore, SHP-1 activity seems to be a critical regulator of oligodendrocyte gene expression and function. Consistent with this role, this study demonstrates that oligodendrocytes of motheaten mice and SHP-1-depleted N20.1 cells produce higher levels of reactive oxygen species (ROS) and exhibit corresponding markers of increased oxidative stress. In agreement with these findings, we demonstrate that increased production of ROS coincides with ROS-induced signaling pathways known to affect myelin gene expression in oligodendrocytes. Antioxidant treatment of SHP-1-deficient oligodendrocytes reversed the pathological changes in these cells, with increased myelin protein gene expression and decreased expression of nuclear factor (erythroid-2)-related factor 2 (Nrf2) responsive gene, heme oxygenase-1 (HO-1). Furthermore, we demonstrate that SHP-1 is expressed in human white matter oligodendrocytes, and there is a subset of multiple sclerosis subjects that demonstrate a deficiency of SHP-1 in normal-appearing white matter. These studies reveal critical pathways controlled by SHP-1 in oligodendrocytes that relate to susceptibility of SHP-1-deficient mice to both developmental defects in myelination and to inflammatory demyelinating diseases.


Subject(s)
Central Nervous System/pathology , Gene Expression Regulation/genetics , Multiple Sclerosis/pathology , Oligodendroglia/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/genetics , Myelin Proteins/genetics , Myelin Proteins/metabolism , NF-kappa B/metabolism , Protein Carbonylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics
4.
Exp Mol Pathol ; 96(3): 393-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24768588

ABSTRACT

Sarcoidosis is an immune-mediated multisystem disease characterized by the formation of non-caseating granulomas. The pathogenesis of sarcoidosis is unclear, with proposed infectious or environmental antigens triggering an aberrant immune response in susceptible hosts. Multiple pro-inflammatory signaling pathways have been implicated in mediating macrophage activation and granuloma formation in sarcoidosis, including IFN-γ/STAT-1, IL-6/STAT-3, and NF-κB. It is difficult to distinguish sarcoidosis from other granulomatous diseases or assess disease severity and treatment response with histopathology alone. Therefore, development of improved diagnostic tools is imperative. Herein, we describe an efficient and reliable technique to classify granulomatous disease through selected gene expression and identify novel genes and cytokine pathways contributing to the pathogenesis of sarcoidosis. We quantified the expression of twenty selected mRNAs extracted from formalin-fixed paraffin embedded (FFPE) tissue (n = 38) of normal lung, suture granulomas, sarcoid granulomas, and fungal granulomas. Utilizing quantitative real-time RT-PCR we analyzed the expression of several genes, including IL-6, COX-2, MCP-1, IFN-γ, T-bet, IRF-1, Nox2, IL-33, and eotaxin-1 and revealed differential regulation between suture, sarcoidosis, and fungal granulomas. This is the first study demonstrating that quantification of target gene expression in FFPE tissue biopsies is a potentially effective diagnostic and research tool in sarcoidosis.


Subject(s)
Genetic Markers , Granuloma/genetics , Sarcoidosis/diagnosis , Sarcoidosis/genetics , Transcriptome , Adolescent , Adult , Aged , Aged, 80 and over , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Child , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gene Expression , Granuloma/immunology , Granuloma/pathology , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-33 , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/genetics , Interleukins/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Middle Aged , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Sarcoidosis/immunology , Sarcoidosis/pathology , Specimen Handling , Up-Regulation , Young Adult
5.
Viruses ; 16(6)2024 May 22.
Article in English | MEDLINE | ID: mdl-38932113

ABSTRACT

Tick-borne flaviviruses (TBFV) can cause severe neuroinvasive disease which may result in death or long-term neurological deficit in over 50% of survivors. Multiple mechanisms for invasion of the central nervous system (CNS) by flaviviruses have been proposed including axonal transport, transcytosis, endothelial infection, and Trojan horse routes. Flaviviruses may utilize different or multiple mechanisms of neuroinvasion depending on the specific virus, infection site, and host variability. In this work we have shown that the infection of BALB/cJ mice with either Powassan virus lineage I (Powassan virus) or lineage II (deer tick virus) results in distinct spatial tropism of infection in the CNS which correlates with unique clinical presentations for each lineage. Comparative transcriptomics of infected brains demonstrates the activation of different immune pathways and downstream host responses. Ultimately, the comparative pathology and transcriptomics are congruent with different clinical signs in a murine model. These results suggest that the different disease presentations occur in clinical cases due to the inherent differences in the two lineages of Powassan virus.


Subject(s)
Brain , Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Mice, Inbred BALB C , Animals , Mice , Encephalitis Viruses, Tick-Borne/pathogenicity , Encephalitis Viruses, Tick-Borne/physiology , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis, Tick-Borne/virology , Encephalitis, Tick-Borne/pathology , Brain/virology , Brain/pathology , Inflammation/virology , Disease Models, Animal , Female , Transcriptome
6.
Psychiatry Res ; 340: 116141, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153291

ABSTRACT

Increased activation of inflammatory macrophages and altered expression of dopamine markers are found in the midbrains of people with schizophrenia (SZ). The relationship of midbrain macrophages to dopamine neurons has not been explored, nor is it known if changes in midbrain macrophages are also present in bipolar disorder (BD) or major depressive disorder (MDD). Herein, we determined whether there were differences in CD163+ cell density in the Substantia Nigra (SN), and cerebral peduncles (CP) of SZ, BD, and MDD compared to controls (CTRL). We also analyzed whether CD163 protein and dopamine-synthesizing enzyme tyrosine hydroxylase (TH) mRNA levels differed among diagnostic groups and if they correlated with the density of macrophages. Overall, perivascular CD163+ cell density was higher in the gray matter (SN) than in the white matter (CP). Compared to CTRL, we found increased density of parenchymal CD163+ cells in the SN of the three psychiatric groups and increased CD163 protein levels in SZ. CD163 protein was positively correlated with density of perivascular CD163+ cells. TH mRNA was reduced in SZ and BD and negatively correlated with parenchymal CD163+ cell density. We provide the first quantitative and molecular evidence of an increase in the density of parenchymal macrophages in the midbrain of major mental illnesses and show that the presence of these macrophages may negatively impact dopaminergic neurons.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Bipolar Disorder , Macrophages , RNA, Messenger , Receptors, Cell Surface , Schizophrenia , Substantia Nigra , Tyrosine 3-Monooxygenase , Humans , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenia/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Male , Macrophages/metabolism , RNA, Messenger/metabolism , Female , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics , Adult , Antigens, CD/metabolism , Middle Aged , Antigens, Differentiation, Myelomonocytic/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Gray Matter/pathology , Gray Matter/metabolism , White Matter/pathology , White Matter/metabolism
7.
Elife ; 122023 05 02.
Article in English | MEDLINE | ID: mdl-37129366

ABSTRACT

Mitochondrial biogenesis requires the import of >1,000 mitochondrial preproteins from the cytosol. Most studies on mitochondrial protein import are focused on the core import machinery. Whether and how the biophysical properties of substrate preproteins affect overall import efficiency is underexplored. Here, we show that protein traffic into mitochondria can be disrupted by amino acid substitutions in a single substrate preprotein. Pathogenic missense mutations in ADP/ATP translocase 1 (ANT1), and its yeast homolog ADP/ATP carrier 2 (Aac2), cause the protein to accumulate along the protein import pathway, thereby obstructing general protein translocation into mitochondria. This impairs mitochondrial respiration, cytosolic proteostasis, and cell viability independent of ANT1's nucleotide transport activity. The mutations act synergistically, as double mutant Aac2/ANT1 causes severe clogging primarily at the translocase of the outer membrane (TOM) complex. This confers extreme toxicity in yeast. In mice, expression of a super-clogger ANT1 variant led to neurodegeneration and an age-dependent dominant myopathy that phenocopy ANT1-induced human disease, suggesting clogging as a mechanism of disease. More broadly, this work implies the existence of uncharacterized amino acid requirements for mitochondrial carrier proteins to avoid clogging and subsequent disease.


Inside our cells, compartments known as mitochondria generate the chemical energy required for life processes to unfold. Most of the proteins found within mitochondria are manufactured in another part of the cell (known as the cytosol) and then imported with the help of specialist machinery. For example, the TOM and TIM22 channels provide a route for the proteins to cross the two membrane barriers that separate the cytosol from the inside of a mitochondrion. ANT1 is a protein that is found inside mitochondria in humans, where it acts as a transport system for the cell's energy currency. Specific mutations in the gene encoding ANT1 have been linked to degenerative conditions that affect the muscles and the brain. However, it remains unclear how these mutations cause disease. To address this question, Coyne et al. recreated some of the mutations in the gene encoding the yeast equivalent of ANT1 (known as Aac2). Experiments in yeast cells carrying these mutations showed that the Aac2 protein accumulated in the TOM and TIM22 channels, creating a 'clog' that prevented other essential proteins from reaching the mitochondria. As a result, the yeast cells died. Mutant forms of the human ANT1 protein also clogged up the TOM and TIM22 channels of human cells in a similar way. Further experiments focused on mice genetically engineered to produce a "super-clogger" version of the mouse equivalent of ANT1. The animals soon developed muscle and neurological conditions similar to those observed in human diseases associated with ANT1. The findings of Coyne et al. suggest that certain genetic mutations in the gene encoding the ANT1 protein cause disease by blocking the transport of other proteins to the mitochondria, rather than by directly affecting ANT1's nucleotide trnsport role in the cell. This redefines our understanding of diseases associated with mitochondrial proteins, potentially altering how treatments for these conditions are designed.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Animals , Humans , Mice , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Carrier Proteins/metabolism , Protein Transport , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism
8.
Clin Immunol ; 142(3): 308-19, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22189043

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS). Here we document for the first time that the cytokine IL-33 is upregulated in both the periphery and the CNS of MS patients. Plasma IL-33 was elevated in MS patients compared to normal subjects and a three-month treatment of MS patients with interferon ß-1a resulted in a significant decrease of IL-33 levels. Similarly, stimulated cultured lymphocytes and macrophages from MS patients had elevated IL-33 levels compared to normal subjects. In parallel, the transcription factor NF-κB that mediates IL-33 transcription was also elevated in leukocytes of MS patients. IL-33 was elevated in normal-appearing white matter and plaque areas from MS brains and astrocytes were identified as an important source of IL-33 expression in the CNS. In summary, IL-33 levels are elevated in the periphery and CNS of MS patients, implicating IL-33 in the pathogenesis of MS.


Subject(s)
Central Nervous System/immunology , Interleukins/immunology , Lymphocytes/immunology , Multiple Sclerosis/immunology , Adult , Cells, Cultured , Female , Humans , Interleukin-33 , Macrophages/immunology , Male , Middle Aged , NF-kappa B/immunology , Up-Regulation
9.
J Vis Exp ; (179)2022 01 21.
Article in English | MEDLINE | ID: mdl-35129175

ABSTRACT

The histologic analysis of brain and spinal cord specimens isolated from mice is common practice for the assessment of pathology in this model system. To maintain the morphology of these delicate tissues, it is routine to administer a chemical fixative such as paraformaldehyde via cannulation of the heart in anesthetized animals (transcardial perfusion). Transcardial perfusion of the mouse heart has traditionally relied on the use of peristaltic pumps or air pressure to deliver both the saline and fixative solutions necessary for this process. As an easily accessible alternative to these methods, this work demonstrates the use of a gravity-fed method of perfusate delivery that uses materials available in most hardware stores. To validate this new perfusion method, this work demonstrates all the subsequent steps necessary for the sensitive detection of phosphorylated α-synuclein in both the brain and spinal cord. Included in these steps are the dissection of the fixed brain and spinal cord tissues, rapid freezing/embedding and cryosectioning of the tissues, and immunofluorescent staining. As this method results in whole-body delivery of the fixative, it may also be used to prepare other non-neuronal tissues for histologic analysis.


Subject(s)
Brain , Spinal Cord , Animals , Brain/pathology , Fixatives , Mice , Perfusion/methods , Spinal Cord/surgery , Staining and Labeling
10.
Front Neurosci ; 16: 858989, 2022.
Article in English | MEDLINE | ID: mdl-35844224

ABSTRACT

Approximately 40% of people with schizophrenia are classified as having "high inflammation." This subgroup has worse neuropathology than patients with "low inflammation." Thus, one would expect the resident microglia and possibly monocyte-derived macrophages infiltrating from the periphery to be "activated" in those with schizophrenia with elevated neuroinflammation. To test whether microglia and/or macrophages are associated with increased inflammatory signaling in schizophrenia, we measured microglia- and macrophage-associated transcripts in the postmortem dorsolateral prefrontal cortex of 69 controls and 72 people with schizophrenia. Both groups were stratified by neuroinflammatory status based on cortical mRNA levels of cytokines and SERPINA3. We found microglial mRNAs levels were either unchanged (IBA1 and Hexb, p > 0.20) or decreased (CD11c, <62% p < 0.001) in high inflammation schizophrenia compared to controls. Conversely, macrophage CD163 mRNA levels were increased in patients, substantially so in the high inflammation schizophrenia subgroup compared to low inflammation subgroup (>250%, p < 0.0001). In contrast, high inflammation controls did not have elevated CD163 mRNA compared to low inflammation controls (p > 0.05). The pro-inflammatory macrophage marker (CD64 mRNA) was elevated (>160%, all p < 0.05) and more related to CD163 mRNA in the high inflammation schizophrenia subgroup compared to high inflammation controls, while anti-inflammatory macrophage and cytokine markers (CD206 and IL-10 mRNAs) were either unchanged or decreased in schizophrenia. Finally, macrophage recruitment chemokine CCL2 mRNA was increased in schizophrenia (>200%, p < 0.0001) and CCL2 mRNA levels positively correlated with CD163 mRNA (r = 0.46, p < 0.0001). Collectively, our findings support the co-existence of quiescent microglia and increased pro-inflammatory macrophages in the cortex of people with schizophrenia.

11.
J Virol ; 83(2): 522-39, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18987138

ABSTRACT

The protein tyrosine phosphatase SHP-1 is a crucial negative regulator of cytokine signaling and inflammatory gene expression, both in the immune system and in the central nervous system (CNS). Mice genetically lacking SHP-1 (me/me) display severe inflammatory demyelinating disease following inoculation with the Theiler's murine encephalomyelitis virus (TMEV) compared to infected wild-type mice. Therefore, it became essential to investigate the mechanisms of TMEV-induced inflammation in the CNS of SHP-1-deficient mice. Herein, we show that the expression of several genes relevant to inflammatory demyelination in the CNS of infected me/me mice is elevated compared to that in wild-type mice. Furthermore, SHP-1 deficiency led to an abundant and exclusive increase in the infiltration of high-level-CD45-expressing (CD45(hi)) CD11b(+) Ly-6C(hi) macrophages into the CNS of me/me mice, in concert with the development of paralysis. Histological analyses of spinal cords revealed the localization of these macrophages to extensive inflammatory demyelinating lesions in infected SHP-1-deficient mice. Sorted populations of CNS-infiltrating macrophages from infected me/me mice showed increased amounts of viral RNA and an enhanced inflammatory profile compared to wild-type macrophages. Importantly, the application of clodronate liposomes effectively depleted splenic and CNS-infiltrating macrophages and significantly delayed the onset of TMEV-induced paralysis. Furthermore, macrophage depletion resulted in lower viral loads and lower levels of inflammatory gene expression and demyelination in the spinal cords of me/me mice. Finally, me/me macrophages were more responsive than wild-type macrophages to chemoattractive stimuli secreted by me/me glial cells, indicating a mechanism for the increased numbers of infiltrating macrophages seen in the CNS of me/me mice. Taken together, these findings demonstrate that infiltrating macrophages in SHP-1-deficient mice play a crucial role in promoting viral replication by providing abundant viral targets and contribute to increased proinflammatory gene expression relevant to the effector mechanisms of macrophage-mediated demyelination.


Subject(s)
Central Nervous System/immunology , Inflammation/immunology , Macrophages/immunology , Poliomyelitis/immunology , Poliomyelitis/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Theilovirus/immunology , Animals , Antigens, Ly/analysis , CD11b Antigen/analysis , Clodronic Acid/pharmacology , Gene Expression Profiling , Immunologic Factors , Leukocyte Common Antigens/analysis , Leukocyte Reduction Procedures , Macrophages/chemistry , Macrophages/virology , Mice , Mice, Inbred C3H , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Spinal Cord/pathology
12.
Lab Invest ; 89(7): 742-59, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398961

ABSTRACT

Recent studies in mice have demonstrated that the protein tyrosine phosphatase SHP-1 is a crucial negative regulator of proinflammatory cytokine signaling, TLR signaling, and inflammatory gene expression. Furthermore, mice genetically lacking SHP-1 (me/me) display a profound susceptibility to inflammatory CNS demyelination relative to wild-type mice. In particular, SHP-1 deficiency may act predominantly in inflammatory macrophages to increase CNS demyelination as SHP-1-deficient macrophages display coexpression of inflammatory effector molecules and increased demyelinating activity in me/me mice. Recently, we reported that PBMCs of multiple sclerosis (MS) patients have a deficiency in SHP-1 expression relative to normal control subjects indicating that SHP-1 deficiency may play a similar role in MS as to that seen in mice. Therefore, it became essential to examine the specific expression and function of SHP-1 in macrophages from MS patients. Herein, we document that macrophages of MS patients have deficient SHP-1 protein and mRNA expression relative to those of normal control subjects. To examine functional consequences of the lower SHP-1, the activation of STAT6, STAT1, and NF-kappaB was quantified and macrophages of MS patients showed increased activation of these transcription factors. In accordance with this observation, several STAT6-, STAT1-, and NF-kappaB-responsive genes that mediate inflammatory demyelination were increased in macrophages of MS patients following cytokine and TLR agonist stimulation. Supporting a direct role of SHP-1 deficiency in altered macrophage function, experimental depletion of SHP-1 in normal subject macrophages resulted in an increased STAT/NF-kappaB activation and increased inflammatory gene expression to levels seen in macrophages of MS patients. In conclusion, macrophages of MS patients display a deficiency of SHP-1 expression, heightened activation of STAT6, STAT1, and NF-kappaB and a corresponding inflammatory profile that may be important in controlling macrophage-mediated demyelination in MS.


Subject(s)
Macrophages/enzymology , Multiple Sclerosis, Relapsing-Remitting/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Adult , Base Sequence , Case-Control Studies , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , DNA Primers/genetics , Demyelinating Diseases/enzymology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Female , Gene Expression , Humans , In Vitro Techniques , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/pathology , NF-kappa B/metabolism , Phenotype , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism
13.
Clin Immunol ; 133(1): 27-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19559654

ABSTRACT

Interferon-beta is a current treatment for multiple sclerosis (MS). Interferon-beta is thought to exert its therapeutic effects on MS by down-modulating the immune response by multiple potential pathways. Here, we document that treatment of MS patients with interferon beta-1a (Rebif) results in a significant increase in the levels and function of the protein tyrosine phosphatase SHP-1 in PBMCs. SHP-1 is a crucial negative regulator of cytokine signaling, inflammatory gene expression, and CNS demyelination as evidenced in mice deficient in SHP-1. In order to examine the functional significance of SHP-1 induction in MS PBMCs, we analyzed the activity of proinflammatory signaling molecules STAT1, STAT6, and NF-kappaB, which are known SHP-1 targets. Interferon-beta treatment in vivo resulted in decreased NF-kappaB and STAT6 activation and increased STAT1 activation. Further analysis in vitro showed that cultured PBMCs of MS patients and normal subjects had a significant SHP-1 induction following interferon-beta treatment that correlated with decreased NF-kappaB and STAT6 activation. Most importantly, experimental depletion of SHP-1 in cultured PBMCs abolished the anti-inflammatory effects of interferon-beta treatment, indicating that SHP-1 is a predominant mediator of interferon-beta activity. In conclusion, interferon-beta treatment upregulates SHP-1 expression resulting in decreased transcription factor activation and inflammatory gene expression important in MS pathogenesis.


Subject(s)
Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/blood , Female , Gene Silencing/immunology , Humans , Interferon beta-1a , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , Multiple Sclerosis/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/immunology , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
J Leukoc Biol ; 84(3): 631-43, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18552204

ABSTRACT

IL-33 is a novel member of the IL-1 cytokine family and a potent inducer of type 2 immunity, as mast cells and Th2 CD4+ T cells respond to IL-33 with the induction of type 2 cytokines such as IL-13. IL-33 mRNA levels are extremely high in the CNS, and CNS glia possess both subunits of the IL-33R, yet whether IL-33 is produced by and affects CNS glia has not been studied. Here, we demonstrate that pathogen-associated molecular patterns (PAMPs) significantly increase IL-33 mRNA and protein expression in CNS glia. Interestingly, IL-33 was localized to the nucleus of astrocytes. Further, CNS glial and astrocyte-enriched cultures treated with a PAMP followed by an ATP pulse had significantly higher levels of supernatant IL-1beta and IL-33 than cultures receiving any single treatment (PAMP or ATP). Supernatants from PAMP + ATP-treated glia induced the secretion of IL-6, IL-13, and MCP-1 from the MC/9 mast cell line in a manner similar to exogenous recombinant IL-33. Further, IL-33 levels and activity were increased in the brains of mice infected with the neurotropic virus Theiler's murine encephalomyelitis virus. IL-33 also had direct effects on CNS glia, as IL-33 induced various innate immune effectors in CNS glia, and this induction was greatly amplified by IL-33-stimulated mast cells. In conclusion, these results implicate IL-33-producing astrocytes as a potentially critical regulator of innate immune responses in the CNS.


Subject(s)
Brain/metabolism , Cardiovirus Infections/metabolism , Interleukins/genetics , Neuroglia/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/immunology , Brain/virology , Cardiovirus Infections/virology , Cell Nucleus/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Immunoenzyme Techniques , Interleukin-1beta , Interleukin-33 , Interleukins/metabolism , Mast Cells/metabolism , Mast Cells/virology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neuroglia/virology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Theilovirus/genetics , Theilovirus/metabolism
15.
J Neuroimmunol ; 331: 46-57, 2019 06 15.
Article in English | MEDLINE | ID: mdl-29113698

ABSTRACT

Glycolysis and mitochondrial respiration are essential for oligodendrocyte metabolism in both the developing and adult CNS. Based on recent reports on the effects of the proinflammatory cytokine IFN-γ on metabolism and on oligodendrocytes, we addressed whether IFN-γ may affect oligodendrocyte bioenergetics in ways relevant to CNS disease. Oligodendrocytes of mice treated with IFN-γ showed significant reductions in aerobic glycolysis and mitochondrial respiration. As expected, IFN-γ treatment led to the induction of STAT1 in oligodendrocytes indicating active signaling into these cells. To determine the direct effects of IFN-γ on oligodendrocyte metabolism, cultured oligodendrocytes were treated with IFN-γ in vitro, which resulted in suppression of glycolysis similar to oligodendrocytes of animals treated with IFN-γ in vivo. Mice lacking SHP-1, a key regulator of IFN-γ and STAT1 signaling in CNS glia, had high constitutive levels of STAT1 and decreased aerobic glycolysis and mitochondrial respiration rates relative to wild type mouse oligodendrocytes. Together, these data show that IFN-γ and SHP-1 control oligodendrocyte bioenergetics in ways that may relate to the role of this cytokine in CNS disease.


Subject(s)
Energy Metabolism/drug effects , Interferon-gamma/pharmacology , Oligodendroglia/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Animals , Cells, Cultured , Central Nervous System/pathology , Enzyme Activation/drug effects , Enzyme Induction/physiology , Female , Glycolysis/drug effects , Male , Mice , Mice, Inbred C3H , Mice, Neurologic Mutants , Oligodendroglia/metabolism , Oxidative Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects
16.
Lab Invest ; 88(3): 243-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18209728

ABSTRACT

Recent studies in mice have demonstrated that the protein tyrosine phosphatase SHP-1 is a crucial negative regulator of cytokine signaling, inflammatory gene expression, and demyelination in central nervous system. The present study investigates a possible similar role for SHP-1 in the human disease multiple sclerosis (MS). The levels of SHP-1 protein and mRNA in PBMCs of MS patients were significantly lower compared to normal subjects. Moreover, promoter II transcripts, expressed from one of two known promoters, were selectively deficient in MS patients. To examine functional consequences of the lower SHP-1 in PBMCs of MS patients, we measured the intracellular levels of phosphorylated STAT6 (pSTAT6). As expected, MS patients had significantly higher levels of pSTAT6. Accordingly, siRNA to SHP-1 effectively increased the levels of pSTAT6 in PBMCs of controls to levels equal to MS patients. Additionally, transduction of PBMCs with a lentiviral vector expressing SHP-1 lowered pSTAT6 levels. Finally, multiple STAT6-responsive inflammatory genes were increased in PBMCs of MS patients relative to PBMCs of normal subjects. Thus, PBMCs of MS patients display a stable deficiency of SHP-1 expression, heightened STAT6 phosphorylation, and an enhanced state of activation relevant to the mechanisms of inflammatory demyelination.


Subject(s)
Gene Expression , Inflammation , Leukocytes, Mononuclear/metabolism , Multiple Sclerosis/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Arginase/analysis , Case-Control Studies , Cells, Cultured , Genetic Vectors , Humans , Lentivirus/genetics , Leukocytes, Mononuclear/drug effects , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , STAT6 Transcription Factor/analysis , STAT6 Transcription Factor/metabolism , Statistics as Topic , Time Factors
17.
J Neurochem ; 105(6): 2511-23, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18331586

ABSTRACT

We have previously shown that the protein tyrosine phosphatase SHP-1 is highly expressed in CNS glia and is an important modulator of cytokine signaling. As such, mice genetically lacking SHP-1 display constitutive myelin abnormalities, severe virus-induced demyelinating disease, and defects in innate anti-viral responses in the CNS. In this study, we show the differential distribution of the SHP-1 promoter-specific transcripts and demonstrate that several cytokines significantly induce SHP-1 expression in CNS glia. Consistent with these cytokine effects, infection with a neurotropic virus both in vitro and in vivo up-regulates SHP-1 transcripts and protein in CNS cells. Using CNS glial cultures of gene knockout mice, we show that interferons-beta and interferons-gamma act through STAT-1 and interferon regulatory factor-1 to induce the SHP-1 promoter I transcripts. Conversely, interferons-beta and IL-6 act through STAT-3 to induce SHP-1 promoter II transcripts. This study demonstrates that interferons and other cytokines associated with virus infections in the CNS can significantly induce the expression of SHP-1 through STAT-1/3 activity and provides a better understanding of the molecular mechanisms regulating cytokine-induced expression important for multiple homeostatic functions of SHP-1 in the CNS.


Subject(s)
Cardiovirus Infections/enzymology , Cytokines/physiology , Encephalitis, Viral/enzymology , Neuroglia/enzymology , Promoter Regions, Genetic/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/biosynthesis , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cells, Cultured , Encephalitis, Viral/genetics , Encephalitis, Viral/metabolism , Enzyme Induction/physiology , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Viral/physiology , Mice , Mice, Inbred C3H , Mice, Knockout , Neuroglia/virology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Theilovirus
18.
J Neuroimmunol ; 122(1-2): 74-84, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11777545

ABSTRACT

Recent studies have shown that the role of the transcription factor interferon regulatory factor-1 (IRF-1) in the expression of major histocompatibility complex (MHC) class I molecules is tissue-specific. Our previous studies indicated a role for IRF-1 in expression of MHC class I genes in cultured astrocytes in response to interferon-gamma (IFN-gamma). However, the requirement for IRF-1 in MHC class I expression has not been directly analyzed in neural tissue. To further ascertain the importance of IRF-1 in the induction of MHC class I genes in astrocytes in response to IFN-gamma, we analyzed astrocytes from mice with a targeted disruption of the IRF-1 gene (IRF-1(-/-) mice). As expected, astrocytes from wild type (IRF-1(+/+)) mice showed a coordinate increase in both IRF-1 and MHC class I gene expression in response to IFN-gamma. To the contrary, astrocytes from IRF-1(-/-) mice had greatly reduced MHC class I mRNA expression. MHC class I gene promoter activity in astrocytes was controlled entirely through a single enhancer, the MHC-IRF-E, to which IRF-1 bound in response to IFN-gamma in wild type but not in IRF-1(-/-) mouse astrocytes. In vivo, astrocytes in brains of wild type mice readily responded to IFN-gamma to express MHC class I molecules. This correlated with increased MHC class I mRNA in the brain. In contrast, brains of IRF-1(-/-) mice showed no MHC class I gene induction following exposure to IFN-gamma indicating that all cells in the central nervous system (CNS) including astrocytes with the potential to express MHC class I molecules were dependent on IRF-1. These studies conclusively demonstrate a major role for IRF-1/MHC-IRF-E interactions in controlling MHC class I gene expression in astrocytes in response to IFN-gamma.


Subject(s)
Antiviral Agents/pharmacology , Astrocytes/immunology , DNA-Binding Proteins/genetics , Histocompatibility Antigens Class I/genetics , Interferon-gamma/pharmacology , Phosphoproteins/genetics , Animals , Brain/cytology , Brain/immunology , Cells, Cultured , DNA-Binding Proteins/immunology , Enhancer Elements, Genetic/immunology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/immunology , Interferon Regulatory Factor-1 , Mice , NF-kappa B/metabolism , Phosphoproteins/immunology , Pregnancy , Promoter Regions, Genetic/immunology , RNA, Messenger/analysis , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
19.
Inflamm Bowel Dis ; 18(12): 2342-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22467146

ABSTRACT

BACKGROUND: Cytokine signaling pathways play a central role in the pathogenesis of inflammatory bowel disease (IBD). Ulcerative colitis (UC) and Crohn's disease (CD) have unique as well as overlapping phenotypes, susceptibility genes, and gene expression profiles. This study aimed to delineate patterns within cytokine signaling pathways in colonic mucosa of UC and CD patients, explore molecular diagnostic markers, and identify novel immune mediators in IBD pathogenesis. METHODS: We quantified 70 selected immune genes that are important in IBD signaling from formalin-fixed, paraffin-embedded (FFPE) colon biopsy samples from normal control subjects and UC and CD patients having either severe colitis or quiescent disease (n = 98 subjects). We utilized and validated a new modified real-time reverse-transcription polymerase chain reaction (RT-PCR) technique for gene quantification. RESULTS: Expression levels of signaling molecules including IL-6/10/12/13/17/23/33, STAT1/3/6, T-bet, GATA3, Foxp3, SOCS1/3, and downstream inflammatory mediators such as chemokines CCL-2/11/17/20, oxidative stress inducers, proteases, and mucosal genes were differentially regulated between UC and CD and between active and quiescent disease. We also document the possible role of novel genes in IBD, including SHP-1, IRF-1,TARC, Eotaxin, NOX2, arginase I, and ADAM 8. CONCLUSIONS: This comprehensive approach to quantifying gene expression provides insights into the pathogenesis of IBD by elucidating distinct immune signaling networks in CD and UC. Furthermore, this is the first study demonstrating that gene expression profiling in FFPE colon biopsies might be a practical and effective tool in the diagnosis and prognosis of IBD and may help identify molecular markers that can predict and monitor response to individualized therapeutic treatments.


Subject(s)
Colitis, Ulcerative/etiology , Crohn Disease/etiology , Cytokines/physiology , Signal Transduction/immunology , Adult , Biomarkers/metabolism , Chemokines/physiology , Colitis, Ulcerative/immunology , Colon/immunology , Crohn Disease/immunology , Female , Genes/immunology , Humans , Intestinal Mucosa/immunology , Male , Oxidative Stress/immunology , Oxidative Stress/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/physiology
20.
J Neuroimmunol ; 246(1-2): 51-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22458980

ABSTRACT

The protein tyrosine phosphatase, SHP-1, is a negative regulator of proinflammatory signaling and autoimmune disease. We have previously reported reduced SHP-1 expression in peripheral blood leukocytes of subjects with multiple sclerosis (MS). Recent evidence indicates that virus-induced DNA methylation of the SHP-1 promoter is responsible for aberrant silencing of SHP-1 expression and function in hematopoietic cells that might relate to inflammatory diseases. In the present study, bisulfite sequencing of the SHP-1 promoter demonstrated that over a third of MS subjects had abnormally high promoter methylation. As SHP-1 is deficient in MS leukocytes and SHP-1-regulated proinflammatory genes are correspondingly upregulated, we propose that increased SHP-1 promoter methylation may relate in part to decreased SHP-1 expression and increased leukocyte-mediated inflammation in MS.


Subject(s)
DNA Methylation/immunology , Leukocytes/immunology , Multiple Sclerosis/immunology , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Up-Regulation/immunology , DNA Methylation/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/blood , Inflammation Mediators/physiology , Leukocytes/metabolism , Leukocytes/pathology , Multiple Sclerosis/blood , Multiple Sclerosis/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL