Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Brain Stimul ; 16(2): 484-506, 2023.
Article in English | MEDLINE | ID: mdl-36773779

ABSTRACT

Vagal fibers travel inside fascicles and form branches to innervate organs and regulate organ functions. Existing vagus nerve stimulation (VNS) therapies activate vagal fibers non-selectively, often resulting in reduced efficacy and side effects from non-targeted organs. The transverse and longitudinal arrangement of fibers inside the vagal trunk with respect to the functions they mediate and organs they innervate is unknown, however it is crucial for selective VNS. Using micro-computed tomography imaging, we tracked fascicular trajectories and found that, in swine, sensory and motor fascicles are spatially separated cephalad, close to the nodose ganglion, and merge caudad, towards the lower cervical and upper thoracic region; larynx-, heart- and lung-specific fascicles are separated caudad and progressively merge cephalad. Using quantified immunohistochemistry at single fiber level, we identified and characterized all vagal fibers and found that fibers of different morphological types are differentially distributed in fascicles: myelinated afferents and efferents occupy separate fascicles, myelinated and unmyelinated efferents also occupy separate fascicles, and small unmyelinated afferents are widely distributed within most fascicles. We developed a multi-contact cuff electrode to accommodate the fascicular structure of the vagal trunk and used it to deliver fascicle-selective cervical VNS in anesthetized and awake swine. Compound action potentials from distinct fiber types, and physiological responses from different organs, including laryngeal muscle, cough, breathing, and heart rate responses are elicited in a radially asymmetric manner, with consistent angular separations that agree with the documented fascicular organization. These results indicate that fibers in the trunk of the vagus nerve are anatomically organized according to functions they mediate and organs they innervate and can be asymmetrically activated by fascicular cervical VNS.


Subject(s)
Vagus Nerve Stimulation , Animals , Swine , Vagus Nerve Stimulation/methods , X-Ray Microtomography , Vagus Nerve/physiology , Action Potentials , Heart Rate
2.
Elife ; 102021 04 06.
Article in English | MEDLINE | ID: mdl-33821789

ABSTRACT

Vagus nerve stimulation (VNS) suppresses inflammation and autoimmune diseases in preclinical and clinical studies. The underlying molecular, neurological, and anatomical mechanisms have been well characterized using acute electrophysiological stimulation of the vagus. However, there are several unanswered mechanistic questions about the effects of chronic VNS, which require solving numerous technical challenges for a long-term interface with the vagus in mice. Here, we describe a scalable model for long-term VNS in mice developed and validated in four research laboratories. We observed significant heart rate responses for at least 4 weeks in 60-90% of animals. Device implantation did not impair vagus-mediated reflexes. VNS using this implant significantly suppressed TNF levels in endotoxemia. Histological examination of implanted nerves revealed fibrotic encapsulation without axonal pathology. This model may be useful to study the physiology of the vagus and provides a tool to systematically investigate long-term VNS as therapy for chronic diseases modeled in mice.


Subject(s)
Electrodes, Implanted/statistics & numerical data , Mice/physiology , Vagus Nerve Stimulation/instrumentation , Vagus Nerve/physiology , Animals , Electrophysiological Phenomena , Male , Mice, Inbred C57BL , Models, Animal
3.
Neuro Oncol ; 21(6): 800-808, 2019 06 10.
Article in English | MEDLINE | ID: mdl-30726976

ABSTRACT

BACKGROUND: Dysembryoplastic neuroepithelial tumors (DNETs) are uncommon neural tumors presenting most often in children and young adults and associated with intractable seizures. Rare midline neoplasms with similar histological features to those found in DNETs have been described near the septum pellucidum and termed "DNET-like neoplasms of the septum pellucidum." Due to their rarity, these tumors have been described in just a few reports and their genetic alterations sought only in small series. METHODS: We collected 20 of these tumors for a comprehensive study of their clinical, radiological, and pathological features. RNA sequencing or targeted DNA sequencing was undertaken on 18 tumors, and genome-wide DNA methylation profiling was possible with 11 tumors. Published cases (n = 22) were also reviewed for comparative purposes. RESULTS: The commonest presenting symptoms and signs were related to raised intracranial pressure; 40% of cases required cerebrospinal fluid diversion. Epilepsy was seen in approximately one third of cases. All patients had an indolent disease course, despite metastasis within the neuraxis in a few cases. Radiologically, the septum verum/septal nuclei were involved in all cases and are the proposed site of origin for septal DNET (sDNET). Septal DNET showed a high frequency (~80%) of mutations of platelet derived growth factor receptor A (PDGFRA), and alterations in fibroblast growth factor receptor 1 (FGFR1) and neurofibromatosis type 1 (NF1) were also identified. In a genomic DNA methylation analysis alongside other neural tumors, sDNETs formed a separate molecular group. CONCLUSIONS: Genetic alterations that are different from those of cerebral DNETs and a distinct methylome profile support the proposal that sDNET is a distinct disease entity.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Magnetic Resonance Imaging/methods , Mutation , Neoplasms, Neuroepithelial/pathology , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Child , DNA Methylation , Female , Humans , Male , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/metabolism , Prognosis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Survival Rate
4.
Arch Pathol Lab Med ; 142(7): 804-814, 2018 07.
Article in English | MEDLINE | ID: mdl-29775073

ABSTRACT

Diffuse gliomas comprise the bulk of "brain cancer" in adults. The recent update to the 4th edition of the World Health Organization's classification of tumors of the central nervous system reflects an unprecedented change in the landscape of the diagnosis and management of diffuse gliomas that will affect all those involved in the management and care of patients. Of the recently discovered gene alterations, mutations in the Krebs cycle enzymes isocitrate dehydrogenases (IDHs) 1 and 2 have fundamentally changed the way the gliomas are understood and classified. Incorporating information on a few genetic parameters (IDH, ATRX and/or p53, and chromosome 1p19q codeletion), a relatively straightforward diagnostic algorithm has been generated with robust and reproducible results that correlate with patients' survival far better than relying on conventional histology alone. Evidence also supports the conclusion that the vast majority of diffuse gliomas without IDH mutations (IDH-wild-type astrocytomas) behave like IDH-wild-type glioblastomas ("molecular GBM"). Together, these changes reflect a big shift in the practice of diagnostic neuropathology in which tumor risk stratification aligns better with molecular information than histology/grading. The purpose of this review is to provide the readers with a brief synopsis of the changes in the 2016 World Health Organization update with an emphasis on diffuse gliomas and to summarize key gene abnormalities on which these classifications are based. Practical points involved in day-to-day diagnostic workup are also discussed, along with a comparison of the various diagnostic tests, including immunohistochemistry, with an emphasis on targeted next-generation sequencing panel technology as a future universal approach.


Subject(s)
Brain Neoplasms/diagnosis , Glioma/diagnosis , Algorithms , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Mutation , Pathology, Molecular , World Health Organization
6.
J Neuroinflammation ; 12: 72, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25890304

ABSTRACT

BACKGROUND: Clinically significant dysregulation of the insulin-like growth factor (IGF) family proteins occurs in HIV-infected individuals, but the details including whether the deficiencies in IGFs contribute to CNS dysfunction are unknown. METHODS: We measured the levels of IGF1, IGF2, IGFBP1, IGFBP2, and IGF2 receptor (IGF2R) in matching plasma and cerebrospinal fluid (CSF) samples of 107 HIV+ individuals from CNS HIV Antiretroviral Therapy Effects Research (CHARTER) and analyzed their associations with demographic and disease characteristics, as well as levels of several soluble inflammatory mediators (TNFα, IL-6, IL-10, IL-17, IP-10, MCP-1, and progranulin). We also determined whether IGF1 or IGF2 deficiency is associated with HIV-associated neurocognitive disorder (HAND) and whether the levels of soluble IGF2R (an IGF scavenging receptor, which we also have found to be a cofactor for HIV infection in vitro) correlate with HIV viral load (VL). RESULTS: There was a positive correlation between the levels of IGF-binding proteins (IGFBPs) and those of inflammatory mediators: between plasma IGFBP1 and IL-17 (ß coefficient 0.28, P = 0.009), plasma IGFBP2 and IL-6 (ß coefficient 0.209, P = 0.021), CSF IGFBP1 and TNFα (ß coefficient 0.394, P < 0.001), and CSF IGFBP2 and TNF-α (ß coefficient 0.14, P < 0.001). As IGFBPs limit IGF availability, these results suggest that inflammation is a significant factor that modulates IGF protein expression/availability in the setting of HIV infection. However, there was no significant association between HAND and the reduced levels of plasma IGF1, IGF2, or CSF IGF1, suggesting a limited power of our study. Interestingly, plasma IGF1 was significantly reduced in subjects on non-nucleoside reverse transcriptase inhibitor-based antiretroviral therapy (ART) compared to protease inhibitor-based therapy (174.1 ± 59.8 vs. 202.8 ± 47.3 ng/ml, P = 0.008), suggesting a scenario in which ART regimen-related toxicity can contribute to HAND. Plasma IGF2R levels were positively correlated with plasma VL (ß coefficient 0.37, P = 0.021) and inversely correlated with current CD4+ T cell counts (ß coefficient -0.04, P = 0.021), supporting our previous findings in vitro. CONCLUSIONS: Together, these results strongly implicate (1) an inverse relationship between inflammation and IGF growth factor availability and the contribution of IGF deficiencies to HAND and (2) the role of IGF2R in HIV infection and as a surrogate biomarker for HIV VL.


Subject(s)
HIV Infections/blood , HIV Infections/cerebrospinal fluid , Somatomedins/metabolism , Adult , Antiretroviral Therapy, Highly Active/methods , Cognition Disorders/blood , Cognition Disorders/cerebrospinal fluid , Cognition Disorders/etiology , Cohort Studies , Cytokines/blood , Cytokines/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , Female , HIV Infections/complications , HIV Infections/drug therapy , Humans , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor Binding Proteins/cerebrospinal fluid , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Male , Middle Aged , Multivariate Analysis , Neurologic Examination , Neuropsychological Tests , Progranulins , Regression Analysis
7.
PLoS One ; 9(7): e103432, 2014.
Article in English | MEDLINE | ID: mdl-25054228

ABSTRACT

OBJECTIVE: Glioblastoma is the most frequent and malignant form of primary brain tumor with grave prognosis. Mounting evidence supports that chronic inflammation (such as chronic overactivation of IL-1 system) is a crucial event in carcinogenesis and tumor progression. IL-1 also is an important cytokine with species-dependent regulations and roles in CNS cell activation. While much attention is paid to specific anti-tumor immunity, little is known about the role of chronic inflammation/innate immunity in glioma pathogenesis. In this study, we examined whether human astrocytic cells (including malignant gliomas) can produce IL-1 and its role in glioma progression. METHODS: We used a combination of cell culture, real-time PCR, ELISA, western blot, immunocytochemistry, siRNA and plasmid transfection, micro-RNA analysis, angiogenesis (tube formation) assay, and neurotoxicity assay. RESULTS: Glioblastoma cells produced large quantities of IL-1 when activated, resembling macrophages/microglia. The activation signal was provided by IL-1 but not the pathogenic components LPS or poly IC. Glioblastoma cells were highly sensitive to IL-1 stimulation, suggesting its relevance in vivo. In human astrocytes, IL-1ß mRNA was not translated to protein. Plasmid transfection also failed to produce IL-1 protein, suggesting active repression. Suppression of microRNAs that can target IL-1α/ß did not induce IL-1 protein. Glioblastoma IL-1ß processing occurred by the NLRP3 inflammasome, and ATP and nigericin increased IL-1ß processing by upregulating NLRP3 expression, similar to macrophages. RNAi of annexin A2, a protein strongly implicated in glioma progression, prevented IL-1 induction, demonstrating its new role in innate immune activation. IL-1 also activated Stat3, a transcription factor crucial in glioma progression. IL-1 activated glioblastoma-conditioned media enhanced angiogenesis and neurotoxicity. CONCLUSIONS: Our results demonstrate unique, species-dependent immune activation mechanisms involving human astrocytes and astrogliomas. Specifically, the ability to produce IL-1 by glioblastoma cells may confer them a mesenchymal phenotype including increased migratory capacity, unique gene signature and proinflammatory signaling.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Aged , Brain/immunology , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Child , Female , Gene Expression Regulation, Neoplastic , Glioma/immunology , Glioma/pathology , Humans , Interleukin-1beta/immunology , Male , Middle Aged , RNA, Messenger/genetics
8.
PLoS One ; 9(5): e98184, 2014.
Article in English | MEDLINE | ID: mdl-24878635

ABSTRACT

BACKGROUND: Compelling data exist that show that normal levels of progranulin (PGRN) are required for successful CNS aging. PGRN production is also modulated by inflammation and infection, but no data are available on the production and role of PGRN during CNS HIV infection. METHODS: To determine the relationships between PGRN and HIV disease, neurocognition, and inflammation, we analyzed 107 matched CSF and plasma samples from CHARTER, a well-characterized HIV cohort. Levels of PGRN were determined by ELISA and compared to levels of several inflammatory mediators (IFNγ, IL-6, IL-10, IP-10, MCP-1, TNFα, IL-1ß, IL-4 and IL-13), as well as clinical, virologic and demographic parameters. The relationship between HIV infection and PGRN was also examined in HIV-infected primary human microglial cultures. RESULTS: In plasma, PGRN levels correlated with the viral load (VL, p<0.001). In the CSF of subjects with undetectable VL, lower PGRN was associated with neurocognitive impairment (p = 0.046). CSF PGRN correlated with CSF IP-10, TNFα and IL-10, and plasma PGRN correlated with plasma IP-10. In vitro, microglial HIV infection increased PGRN production and PGRN knockdown increased HIV replication, demonstrating that PGRN is an innate antiviral protein. CONCLUSIONS: We propose that PGRN plays dual roles in people living with HIV disease. With active HIV replication, PGRN is induced in infected macrophages and microglia and functions as an antiviral protein. In individuals without active viral replication, decreased PGRN production contributes to neurocognitive dysfunction, probably through a diminution of its neurotrophic functions. Our results have implications for the pathogenesis, biomarker studies and therapy for HIV diseases including HIV-associated neurocognitive dysfunction (HAND).


Subject(s)
Cognition , HIV Infections/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Adult , Female , HIV/physiology , HIV Infections/blood , HIV Infections/cerebrospinal fluid , HIV Infections/physiopathology , Humans , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Male , Microglia/metabolism , Microglia/virology , Multivariate Analysis , Progranulins , Viral Load , Virus Replication
9.
Glia ; 62(6): 999-1013, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659539

ABSTRACT

Treatment of cultures with toll-like receptor (TLR) ligands or cytokines has become a popular approach to investigate astrocyte neuroinflammatory responses and to simulate the neural environment in various CNS disorders. However, despite much effort, the mechanism of astrocyte activation such as their responses to the TLR ligands and IL-1 remain highly debated. We compared highly pure primary mouse and human astrocyte cultures in their ability to produce proinflammatory mediators (termed "A1") and immunoregulatory mediators (termed "A2") in response to LPS, poly IC, and IL-1 stimulation. In human astrocytes, IL-1 induced both A1 and A2 responses, poly IC induced mostly A2, and LPS induced neither. In mouse astrocytes, LPS induced mostly an A1-predominant response, poly IC induced both A1 and A2, and IL-1 neither. In addition, mouse astrocytes produce abundant IL-1 protein, whereas human astrocytes did not, despite robust IL-1 mRNA expression. Of the TLR4 receptor complex proteins, human astrocytes expressed TLR4 and MD2 but not CD14, whereas mouse astrocytes expressed all three. Mouse astrocyte CD14 (cell-associated and soluble) was potently upregulated by LPS. Silencing TLR4 or CD14 by siRNA suppressed LPS responses in mouse astrocytes. In vivo, astrocytes in LPS-injected mouse brains also expressed CD14. Our results show striking differences between human and mouse astrocytes in the use of TLR/IL-1R and subsequent downstream signaling and immune activation. IL-1 translational block in human astrocytes may be a built-in mechanism to prevent autocrine and paracrine cell activation and neuroinflammation. These results have important implications for translational research of human CNS diseases.


Subject(s)
Astrocytes/drug effects , Astrocytes/physiology , Interleukin-1/toxicity , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/toxicity , Animals , Animals, Newborn , Cells, Cultured , Fetus , Humans , Mice , Mice, Inbred C57BL , Species Specificity
10.
J Proteomics ; 99: 152-168, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24503185

ABSTRACT

The tumor microenvironment including glial cells and their inflammatory products regulates brain tumor development and progression. We have previously established that human glioma cells are exquisitely sensitive to IL-1 stimulation leading us to undertake a comparative analysis of the secretome of unstimulated and cytokine (IL-1)-stimulated glioblastoma cells. We performed label-free quantitative proteomic analysis and detected 190 proteins which included cytokines, chemokines, growth factors, proteases, cell adhesion molecules, extracellular matrix (ECM) and related proteins. Measuring area under the curve (AUC) of peptides for quantitation, the IL-1-induced secretome contained 13 upregulated and 5 downregulated extracellular proteins (p<0.05) compared to controls. Of these, IL-8, CCL2, TNC, Gal-1 and PTX3 were validated as upregulated and SERPINE1, STC2, CTGF and COL4A2 were validated as downregulated factors by immunochemical methods. A major representation of the ECM and related proteins in the glioblastoma secretome and their modulation by IL-1 suggested that IL-1 induces its effect in part by altering TGFß expression, activity and signaling. These findings enhance our understanding of IL-1-induced modulation of glioma microenvironment, with implications for increased tumor invasion, migration and angiogenesis. They further provide novel targets for the glioblastoma intervention. BIOLOGICAL SIGNIFICANCE: Present study is on an unbiased screening of the glioblastoma secretome stimulated by IL-1 which triggers neuroinflammatory cascades in the central nervous system. Network of secreted proteins were shown to be regulated revealing their possible contribution to glioma progression. Label free quantitative proteomics has provided unique novel targets for potential glioblastoma intervention.


Subject(s)
Cell Movement , Glioblastoma/metabolism , Interleukin-1/pharmacology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Proteome/metabolism , Tumor Microenvironment , Cell Line, Tumor , Glioblastoma/pathology , Humans , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/pathology
11.
J Neuroimmune Pharmacol ; 9(2): 117-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23959579

ABSTRACT

Progranulin (PGRN) is a highly unusual molecule with both neuronal and microglial expression with two seemingly unrelated functions, i.e., as a neuronal growth factor and a modulator of neuroinflammation. Haploinsufficiency due to loss of function mutations lead to a fatal presenile dementing illness (frontotemporal lobar degeneration), indicating that adequate expression of PGRN is essential for successful aging. PGRN might be a particularly relevant factor in the pathogenesis of HIVencephalitis (HIVE) and HIV-associated neurocognitive disorders (HAND). We present emerging data and a review of the literature which show that cells of myeloid lineage such as macrophages and microglia are the primary sources of PGRN and that PGRN expression contributes to pathogenesis of CNS diseases. We also present evidence that PGRN is a macrophage antiviral cytokine. For example, PGRN mRNA and protein expression are significantly upregulated in brain specimens with HIVE, and in HIV infected microglia in vitro. Paradoxically, our preliminary CHARTER data analyses indicate that lower PGRN levels in CSF trended towards an association with HAND, particularly in those without detectable virus. Based upon these findings, we introduce the hypothesis that PGRN plays dual roles in modulating antiviral immunity and neuronal dysfunction in the context of HIV infection. In the presence of active viral replication, PGRN expression is increased functioning as an anti-viral factor as well as a neuroprotectant. In the absence of active HIV replication, ongoing inflammation or other stressors suppress PGRN production from macrophages/microglia contributing to neurocognitive dysfunction. We propose.


Subject(s)
AIDS Dementia Complex/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Microglia/metabolism , AIDS Dementia Complex/pathology , Animals , Cognition Disorders/metabolism , Cognition Disorders/virology , Humans , Progranulins
12.
PLoS One ; 8(6): e66657, 2013.
Article in English | MEDLINE | ID: mdl-23885286

ABSTRACT

Pannexin1 (Panx1) is a plasma membrane channel permeable to relatively large molecules, such as ATP. In the central nervous system (CNS) Panx1 is found in neurons and glia and in the immune system in macrophages and T-cells. We tested the hypothesis that Panx1-mediated ATP release contributes to expression of Experimental Autoimmune Encephalomyelitis (EAE), an animal model for multiple sclerosis, using wild-type (WT) and Panx1 knockout (KO) mice. Panx1 KO mice displayed a delayed onset of clinical signs of EAE and decreased mortality compared to WT mice, but developed as severe symptoms as the surviving WT mice. Spinal cord inflammatory lesions were also reduced in Panx1 KO EAE mice during acute disease. Additionally, pharmacologic inhibition of Panx1 channels with mefloquine (MFQ) reduced severity of acute and chronic EAE when administered before or after onset of clinical signs. ATP release and YoPro uptake were significantly increased in WT mice with EAE as compared to WT non-EAE and reduced in tissues of EAE Panx1 KO mice. Interestingly, we found that the P2X7 receptor was upregulated in the chronic phase of EAE in both WT and Panx1 KO spinal cords. Such increase in receptor expression is likely to counterbalance the decrease in ATP release recorded from Panx1 KO mice and thus contribute to the development of EAE symptoms in these mice. The present study shows that a Panx1 dependent mechanism (ATP release and/or inflammasome activation) contributes to disease progression, and that inhibition of Panx1 using pharmacology or gene disruption delays and attenuates clinical signs of EAE.


Subject(s)
Connexins/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Nerve Tissue Proteins/physiology , Animals , Base Sequence , Connexins/genetics , DNA Primers , Encephalomyelitis, Autoimmune, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X7/metabolism , Spleen/metabolism , Spleen/pathology
13.
J Neurooncol ; 113(2): 185-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512614

ABSTRACT

Glioblastoma multiforme (GBM) is the most common, highly malignant primary tumor of the brain with poor prognosis. Even with the improved therapy regimen including temozolomide, the average survival rate is less than 2 years. Additional approaches to therapy targeting multiple aspects of glioma progression are in need. In the present work, we have tested the possibility that upregulation of the transcription factor interferon regulatory factor 3 (IRF3) can inhibit glioma invasiveness, proliferation and production of pro-inflammatory and pro-angiogenic factors in cultures of malignant glioma cell lines (U271, U87 and SNB-19). IRF3 is an essential transcription factor involved in TLR3/4-mediated signaling and generation of type I interferons. Although IRF3 has been suggested as a potential tumor suppressor gene, its role in glioma remains uninvestigated. In this study, we find that human glioma immune activation is potently elicited by a cytokine combination, IL-1/IFNγ (or poly IC), but not by bacterial lipopolysaccharide (LPS), similar to primary human astrocytes. GBM biopsy specimens show little detectable IRF3 immunoreactivity, and in vitro adenovirus-mediated IRF3 gene transfer in glioma cells modulates IL-1/IFNγ-induced cytokine and chemokine genes, resulting in upregulation of IFNß and IP-10 (IRF3-stimulated genes) and downregulation of proinflammatory and angiogenic genes including IL-8, TNFα and VEGF (IRF3-represssed genes). Cytokines (IL-1ß and TNFα) also induce the expression of miR-155 and miR-155*, the microRNAs crucial in immunity and inflammation-induced oncogenesis and this is dose-dependently suppressed by IRF3. Importantly, IRF3 also inhibits glioma proliferation, migration and invasion. Together, these data suggest that IRF3 can suppress glioma progression. Agents that promote IRF3 activation and expression (such as IRF3 gene transfer) could be explored as potential future therapy.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Glioma/pathology , Inflammation Mediators/metabolism , Inflammation/pathology , Interferon Regulatory Factor-3/metabolism , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Adhesion , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Wound Healing
14.
J Neuroinflammation ; 10: 37, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23497056

ABSTRACT

BACKGROUND: Recent studies in experimental animals show that insulin-like growth factor 1 (IGF1) plays a trophic role during development and tissue injury and that microglia are important sources of IGF1. However, little information is available regarding the expression, regulation, and function of IGF1 and related proteins in human brain cells. In the current study, we examined the expression of IGF1 and IGF2 in human microglia in vivo and in vitro. METHODS: Expression of IGF1 and IGF2 was examined by immunohistochemistry in post-mortem human brain sections derived from HIV+ and HIV- brains. In primary cultures of human fetal microglia, IGF1 and IGF2 mRNA and protein expression was examined by Q-PCR, ELISA, and Western blot analysis. Additionally, the role of IGF1 and IGF2 in neuroprotection was examined in primary human neuronal glial cultures. RESULTS: Immunohistochemistry of human brain tissues showed that nonparenchymal cells (vessels and meninges), as well as parenchymal microglia and macrophages were positive for IGF1, in both HIV encephalitis and control brains, while IGF2 was undetectable. Cultured microglia expressed IGF1 mRNA and produced pg/ml levels of IGF1 protein; this was significantly suppressed by proinflammatory mediators, such as lipopolysaccharide (LPS), poly(I:C), and IFNγ. The Th2 cytokines IL-4 and IL-13 had no significant effect, but the cAMP analog (dibutyryl cAMP) significantly increased IGF1 production. In contrast, microglial IGF2 mRNA and protein (determined by Western blot) were upregulated by LPS. IGF1 receptor (IGF1R) immunoreactivity was predominantly expressed by neurons, and both IGF1 and IGF2 significantly protected neurons from cytokine (IL-1/IFNγ) induced death. CONCLUSIONS: Our study in human brain tissues and cells indicates that microglia are important sources of neurotrophic growth factors IGF1 and IGF2, and that microglial activation phenotypes can influence the growth factor expression. Importantly, our results suggest that chronic neuroinflammation and upregulation of proinflammatory cytokines could lead to neurodegeneration by suppressing the production of microglia-derived neuronal growth factors, such as IGF1.


Subject(s)
Gene Expression Regulation , Inflammation Mediators/physiology , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor I/biosynthesis , Microglia/metabolism , Cells, Cultured , Coculture Techniques , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/genetics , Microglia/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Phenotype
15.
J Neuroimmune Pharmacol ; 8(1): 132-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22684309

ABSTRACT

Interferon regulatory factor 3 (IRF3) is a transcription factor critical in the induction of antiviral immunity. IRF3 is activated following stimulation of cell membrane or cytosolic nucleic acid sensors and is essential in the induction of the IFNß gene. Most cells constitutively express IRF3 in vitro, but little is known about the regulation of expression of IRF3 in vivo. Immunohistochemical analysis of selected human and mouse tissues demonstrated that IRF3 expression is highly organ- and cell-type specific, showing high expression in certain epithelial cells. In the CNS, while ependymal cells are strongly positive, brain parenchyma has little detectable IRF3 immunoreactivity. The importance of IRF3 in antiviral immunity has been demonstrated by the requirement for IRF3 in suppressing viral replication, but also by the demonstration that virus degrades IRF3 protein in infected cells. Furthermore, HIV-infected microglia in human CNS show abnormal IRF3+ aggregates, indicative of aberrant protein processing in vivo. In addition to antiviral immunity, IRF3 also plays a critical role in the modulation of neuroinflammation. A combination of dominant-negative and over-expression strategies in vitro as well as transgenic expression of IRF3 in vivo demonstrated that IRF3 plays a major role in modulating glial cytokine expression, i.e., suppression of proinflammatory cytokines and promotion of anti-inflammatory or immunoregulatory cytokines. These observations together suggest that IRF3 is a crucial regulator of immune responses against pathogen- and damage-associated molecules. We review recent literature on the molecular pathways of IRF3 activation and function of IRF3 and discuss their implications for CNS diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Antiviral Agents , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/immunology , Interferon Regulatory Factor-3/pharmacology , Animals , Central Nervous System Diseases/prevention & control , Encephalitis/immunology , Encephalitis/pathology , Humans , Interferon Regulatory Factor-3/chemistry
16.
J Neuroinflammation ; 9: 208, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22938163

ABSTRACT

BACKGROUND: Expression of active c-Abl in adult mouse forebrain neurons in the AblPP/tTA mice resulted in severe neurodegeneration, particularly in the CA1 region of the hippocampus. Neuronal loss was preceded and accompanied by substantial microgliosis and astrocytosis. In contrast, expression of constitutively active Arg (Abl-related gene) in mouse forebrain neurons (ArgPP/tTA mice) caused no detectable neuronal loss or gliosis, although protein expression and kinase activity were at similar levels to those in the AblPP/tTA mice. METHODS: To begin to elucidate the mechanism of c-Abl-induced neuronal loss and gliosis, gene expression analysis of AblPP/tTA mouse forebrain prior to development of overt pathology was performed. Selected results from gene expression studies were validated with quantitative reverse transcription PCR , immunoblotting and bromodeoxyuridine (BrdU) labeling, and by immunocytochemistry. RESULTS: Two of the top pathways upregulated in AblPP/tTA mice with c-Abl expression for 2 weeks were cell cycle and interferon signaling. However, only the expression of interferon signaling pathway genes remained elevated at 4 weeks of c-Abl induction. BrdU incorporation studies confirm that, while the cell cycle pathway is upregulated in AblPP/tTA mice at 2 weeks of c-Abl induction, the anatomical localization of the pathway is not consistent with previous pathology seen in the AblPP/tTA mice. Increased expression and activation of STAT1, a known component of interferon signaling and interferon-induced neuronal excitotoxicity, is an early consequence of c-Abl activation in AblPP/tTA mice and occurs in the CA1 region of the hippocampus, the same region that goes on to develop severe neurodegenerative pathology and neuroinflammation. Interestingly, no upregulation of gene expression of interferons themselves was detected. CONCLUSIONS: Our data suggest that the interferon signaling pathway may play a role in the pathologic processes caused by c-Abl expression in neurons, and that the AblPP/tTA mouse may be an excellent model for studying sterile inflammation and the effects of interferon signaling in the brain.


Subject(s)
Cell Cycle/physiology , Interferons/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Doxycycline/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Transgenic , Neurogenesis/genetics , Olfactory Pathways/metabolism , Oncogene Proteins v-abl/genetics , Prosencephalon/cytology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics
17.
PLoS One ; 7(4): e35115, 2012.
Article in English | MEDLINE | ID: mdl-22509390

ABSTRACT

BACKGROUND: The essential role of progranulin (PGRN) as a neurotrophic factor has been demonstrated by the discovery that haploinsufficiency due to GRN gene mutations causes frontotemporal lobar dementia. In addition to neurons, microglia in vivo express PGRN, but little is known about the regulation of PGRN expression by microglia. GOAL: In the current study, we examined the regulation of expression and function of PGRN, its proteolytic enzyme macrophage elastase (MMP-12), as well as the inhibitor of PGRN proteolysis, secretory leukocyte protease inhibitor (SLPI), in human CNS cells. METHODS: Cultures of primary human microglia and astrocytes were stimulated with the TLR ligands (LPS or poly IC), Th1 cytokines (IL-1/IFNγ), or Th2 cytokines (IL-4, IL-13). Results were analyzed by Q-PCR, immunoblotting or ELISA. The roles of MMP-12 and SLPI in PGRN cleavage were also examined. RESULTS: Unstimulated microglia produced nanogram levels of PGRN, and PGRN release from microglia was suppressed by the TLR ligands or IL-1/IFNγ, but increased by IL-4 or IL-13. Unexpectedly, while astrocytes stimulated with proinflammatory factors released large amounts of SLPI, none were detected in microglial cultures. We also identified MMP-12 as a PGRN proteolytic enzyme, and SLPI as an inhibitor of MMP-12-induced PGRN proteolysis. Experiments employing PGRN siRNA demonstrated that microglial PGRN was involved in the cytokine and chemokine production following TLR3/4 activation, with its effect on TNFα being the most conspicuous. CONCLUSIONS: Our study is the first detailed examination of PGRN in human microglia. Our results establish microglia as a significant source of PGRN, and MMP-12 and SLPI as modulators of PGRN proteolysis. Negative and positive regulation of microglial PGRN release by the proinflammatory/Th1 and the Th2 stimuli, respectively, suggests a fundamentally different aspect of PGRN regulation compared to other known microglial activation products. Microglial PGRN appears to function as an endogenous modulator of innate immune responses.


Subject(s)
Astrocytes , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 12/metabolism , Microglia , Astrocytes/enzymology , Astrocytes/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Intercellular Signaling Peptides and Proteins/genetics , Matrix Metalloproteinase 12/genetics , Microglia/enzymology , Microglia/metabolism , Progranulins , Proteolysis , RNA, Small Interfering , Secretory Leukocyte Peptidase Inhibitor/metabolism , Signal Transduction
18.
Glia ; 59(12): 1911-22, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22170100

ABSTRACT

Astrocytes, together with microglia and macrophages, participate in innate inflammatory responses in the CNS. Although inflammatory mediators such as interferons generated by astrocytes may be critical in the defense of the CNS, sustained unopposed cytokine signaling could result in harmful consequences. Interferon regulatory factor 3 (IRF3) is a transcription factor required for IFNß production and antiviral immunity. Most cells express low levels of IRF3 protein, and the transcriptional mechanism that upregulates IRF3 expression is not known. In this study, we explored the consequence of adenovirus-mediated IRF3 gene transfer (Ad-IRF3) in primary human astrocytes. We show that IRF3 transgene expression suppresses proinflammatory cytokine gene expression upon challenge with IL-1/IFNγ and alters astrocyte activation phenotype from a proinflammatory to an anti-inflammatory one, akin to an M1-M2 switch in macrophages. This was accompanied by the rescue of neurons from cytokine-induced death in glial-neuronal co-cultures. Furthermore, Ad-IRF3 suppressed the expression of microRNA-155 and its star-form partner miR-155*, immunoregulatory miRNAs highly expressed in multiple sclerosis lesions. Astrocyte miR-155/miR155* were induced by cytokines and TLR ligands with a distinct hierarchy and involved in proinflammatory cytokine gene induction by targeting suppressor of cytokine signaling 1, a negative regulator of cytokine signaling and potentially other factors. Our results demonstrate a novel proinflammatory role for miR-155/miR-155* in human astrocytes and suggest that IRF3 can suppress neuroinflammation through regulating immunomodulatory miRNA expression. © 2011 Wiley-Liss, Inc.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Gene Expression Regulation/genetics , Interferon Regulatory Factor-3/physiology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Coculture Techniques , Humans , Interferon Regulatory Factor-3/biosynthesis , Interferon Regulatory Factor-3/genetics , Phenotype , Primary Cell Culture
19.
Am J Pathol ; 179(3): 1360-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21855684

ABSTRACT

Tryptophan metabolism by the kynurenine pathway (KP) is important to the pathogenesis of inflammatory, infectious, and degenerative diseases. The 3-hydroxykynurenine (3-HK) branch of the KP is activated in macrophages and microglia, leading to the generation of 3-HK, 3-hydroxyanthranilic acid (3-HAA), and quinolinic acid, which are considered neurotoxic owing to their free radical-generating and N-methyl-d-aspartic acid receptor agonist activities. We investigated the role of 3-HAA in inflammatory and antioxidant gene expression and neurotoxicity in primary human fetal central nervous system cultures treated with cytokines (IL-1 with or without interferon-γ) or with Toll-like receptor ligands mimicking the proinflammatory central nervous system environment. Results were analyzed by microarray, Western blot, immunostain, enzyme-linked immunosorbent assay, and neurotoxicity assays. 3-HAA suppressed glial cytokine and chemokine expression and reduced cytokine-induced neuronal death. 3-HK also suppressed cytokine-induced neuronal death. Unexpectedly, 3-HAA was highly effective in inducing in astrocytes the expression of hemeoxygenase-1 (HO-1), an antioxidant enzyme with anti-inflammatory and cytoprotective properties. Optimal induction of HO-1 required 3-HAA and cytokines. In human microglia, 3-HAA weakly induced HO-1 and lipopolysaccharide suppressed microglial HO-1 expression. 3-HAA-mediated HO-1 expression was confirmed in cultured adult human astrocytes and in vivo after 3-HAA injection to mouse brains. Together, our results demonstrate the novel neuroprotective activity of the tryptophan metabolite 3-HAA and have implications for future therapeutic approaches for neuroinflammatory disorders.


Subject(s)
3-Hydroxyanthranilic Acid/pharmacology , Anti-Inflammatory Agents/pharmacology , Heme Oxygenase-1/metabolism , Nootropic Agents/pharmacology , 3-Hydroxyanthranilic Acid/metabolism , Adult , Animals , Astrocytes/metabolism , Cell Death/drug effects , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Kynurenine/analogs & derivatives , Kynurenine/metabolism , Mice , Microglia/metabolism , Neurons/drug effects
20.
J Neuroinflammation ; 8: 187, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22208359

ABSTRACT

BACKGROUND: Microglia are the principal cells involved in the innate immune response in the CNS. Activated microglia produce a number of proinflammatory cytokines implicated in neurotoxicity but they also are a major source of anti-inflammatory cytokines, antiviral proteins and growth factors. Therefore, an immune therapy aiming at suppressing the proinflammatory phenotype while enhancing the anti-inflammatory, growth promoting phenotype would be of great benefit. In the current study, we tested the hypothesis that interferon regulatory factor 3 (IRF3), a transcription factor required for the induction of IFNß following TLR3 or TLR4 activation, is critical to the microglial phenotype change from proinflammatory to anti-inflammatory, and that this phenotype change can be greatly facilitated by IRF3 gene transfer. METHODS: Cultures of primary human fetal microglia were transduced with IRF3 using recombinant adenovirus (Ad-IRF3) and subjected to microarray analysis, real-time PCR, immunoblotting and ELISA to determine inflammatory gene expression. Two different types of immune stimuli were tested, the TLR ligands, poly IC (PIC) and LPS, and the proinflammatory cytokines, IL-1/IFNγ. In addition, the role of the PI3K/Akt pathway was examined by use of a pharmacological inhibitor, LY294002. RESULTS: Our results show that Ad-IRF3 suppressed proinflammatory genes (IL-1α, IL-1ß, TNFα, IL-6, IL-8 and CXCL1) and enhanced anti-inflammatory genes (IL-1 receptor antagonist, IL-10 and IFNß) in microglia, regardless of the cell stimuli applied. Furthermore, Ad-IRF3 activated Akt, and LY294002 reversed the effects of Ad-IRF3 on microglial inflammatory gene expression. pAkt was critical in LPS- or PIC-induced production of IL-10 and IL-1ra. Significantly, microglial IFNß protein production was also dependent on pAkt and required both Ad-IRF3 and immunological stimuli (PIC > IL-1/IFNγ). pAkt played much less prominent and variable roles in microglial proinflammatory gene expression. This anti-inflammatory promoting role of PI3K/Akt appeared to be specific to microglia, since astrocyte proinflammatory gene expression (as well as IFNß expression) required PI3K/Akt. CONCLUSIONS: Our results show a novel anti-inflammatory role for the PI3K/Akt signaling pathway in microglia. They further suggest that IRF3 gene therapy could facilitate the microglial phenotype switch from proinflammatory ("M1-like") to anti-inflammatory and immunomodulatory ("M2-like"), in part, by augmenting the level of pAkt.


Subject(s)
Interferon Regulatory Factor-3/immunology , Microglia/immunology , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , Cells, Cultured , Chemokines/genetics , Chemokines/immunology , Chromones/pharmacology , Cytokines/genetics , Cytokines/immunology , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Transfer Techniques , Humans , Immunity, Innate/immunology , Interferon Regulatory Factor-3/genetics , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Microarray Analysis , Microglia/cytology , Microglia/drug effects , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...