Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873446

ABSTRACT

In multiple sclerosis (MS), the invasion of the central nervous system by peripheral immune cells is followed by the activation of resident microglia and astrocytes. This cascade of events results in demyelination, which triggers neuronal damage and death. The molecular signals in neurons responsible for this damage are not yet fully characterized. In MS, retinal ganglion cell neurons (RGCs) of the central nervous system (CNS) undergo axonal injury and cell death. This phenomenon is mirrored in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. To understand the molecular landscape, we isolated RGCs from mice subjected to the EAE protocol. RNA-sequencing and ATAC-sequencing analyses were performed. Pathway analysis of the RNA-sequencing data revealed that RGCs displayed a molecular signature, similar to aged neurons, showcasing features of senescence. Single-nucleus RNA-sequencing analysis of neurons from human MS patients revealed a comparable senescence-like phenotype., which was supported by immunostaining RGCs in EAE mice. These changes include alterations to the nuclear envelope, modifications in chromatin marks, and accumulation of DNA damage. Transduction of RGCs with an Oct4 - Sox2 - Klf4 transgene to convert neurons in the EAE model to a more youthful epigenetic and transcriptomic state enhanced the survival of RGCs. Collectively, this research uncovers a previously unidentified senescent-like phenotype in neurons under pathological inflammation and neurons from MS patients. The rejuvenation of this aged transcriptome improved visual acuity and neuronal survival in the EAE model supporting the idea that age rejuvenation therapies and senotherapeutic agents could offer a direct means of neuroprotection in autoimmune disorders.

2.
Nat Immunol ; 24(7): 1188-1199, 2023 07.
Article in English | MEDLINE | ID: mdl-37322178

ABSTRACT

Spalt-like transcription factor 1 (SALL1) is a critical regulator of organogenesis and microglia identity. Here we demonstrate that disruption of a conserved microglia-specific super-enhancer interacting with the Sall1 promoter results in complete and specific loss of Sall1 expression in microglia. By determining the genomic binding sites of SALL1 and leveraging Sall1 enhancer knockout mice, we provide evidence for functional interactions between SALL1 and SMAD4 required for microglia-specific gene expression. SMAD4 binds directly to the Sall1 super-enhancer and is required for Sall1 expression, consistent with an evolutionarily conserved requirement of the TGFß and SMAD homologs Dpp and Mad for cell-specific expression of Spalt in the Drosophila wing. Unexpectedly, SALL1 in turn promotes binding and function of SMAD4 at microglia-specific enhancers while simultaneously suppressing binding of SMAD4 to enhancers of genes that become inappropriately activated in enhancer knockout microglia, thereby enforcing microglia-specific functions of the TGFß-SMAD signaling axis.


Subject(s)
Microglia , Transcription Factors , Animals , Mice , Binding Sites , DNA , Mice, Knockout , Microglia/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
3.
Nat Rev Neurosci ; 24(7): 397-415, 2023 07.
Article in English | MEDLINE | ID: mdl-37268822

ABSTRACT

As resident macrophages of the CNS, microglia are critical immune effectors of inflammatory lesions and associated neural dysfunctions. In multiple sclerosis (MS) and its animal models, chronic microglial inflammatory activity damages myelin and disrupts axonal and synaptic activity. In contrast to these detrimental effects, the potent phagocytic and tissue-remodelling capabilities of microglia support critical endogenous repair mechanisms. Although these opposing capabilities have long been appreciated, a precise understanding of their underlying molecular effectors is only beginning to emerge. Here, we review recent advances in our understanding of the roles of microglia in animal models of MS and demyelinating lesions and the mechanisms that underlie their damaging and repairing activities. We also discuss how the structured organization and regulation of the genome enables complex transcriptional heterogeneity within the microglial cell population at demyelinating lesions.


Subject(s)
Multiple Sclerosis , Animals , Microglia/physiology , Macrophages , Axons/pathology , Inflammation/pathology , Disease Models, Animal
4.
Life Sci Alliance ; 5(10)2022 10.
Article in English | MEDLINE | ID: mdl-35705491

ABSTRACT

Remyelination failure in multiple sclerosis leads to progressive demyelination and inflammation, resulting in neurodegeneration and clinical decline. Microglia are innate immune cells that can acquire a regenerative phenotype to promote remyelination, yet little is known about the regulators controlling the regenerative microglia activation. Herein, using a cuprizone (CPZ)-diet induced de- and remyelination mice model, we identify PRMT1 as a driver for MHC-associated microglia population required for remyelination in the central nervous system. The loss of PRMT1, but not PRMT5, in microglia resulted in impairment of the remyelination with a reduction of oligoprogenitor cell number and prolonged microgliosis and astrogliosis. Using single-cell RNA sequencing, we found eight distinct microglial clusters during the CPZ diet, and PRMT1 depleted microglia hindered the formation of the MHC-associated cluster, expressing MHCII and CD11c. Mechanistically, PRMT1-KO microglia displayed reduced the H3K27ac peaks at the promoter regions of the MHC- and IFN-associated genes and further suppressed gene expression during CPZ diet. Overall, our findings demonstrate that PRMT1 is a critical regulator of the MHC- and IFN-associated microglia, necessary for central nervous system remyelination.


Subject(s)
Demyelinating Diseases , Remyelination , Animals , Central Nervous System/metabolism , Cuprizone/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism
5.
Glia ; 70(3): 572-589, 2022 03.
Article in English | MEDLINE | ID: mdl-34862814

ABSTRACT

Microglia proliferate during brain development and brain lesions, but how this is coordinated at the transcriptional level is not well understood. Here, we investigated fundamental aspects of the transcriptional process associated with proliferation of mouse microglia during postnatal development and in adults in a model of induced microglial depletion-repopulation. While each proliferative subset displayed globally a distinct signature of gene expression, they also co-expressed a subgroup of 1370 genes at higher levels than quiescent microglia. Expression of these may be coordinated by one of two mechanisms of regulation with distinct properties. A first mechanism augments expression of genes already expressed in quiescent microglia and is subject to regulation by Klf/Sp, Nfy, and Ets transcription factors. Alternatively, a second mechanism enables de novo transcription of cell cycle genes and requires additional regulatory input from Lin54 and E2f transcription factors. Of note, transcriptional upregulation of E2f1 and E2f2 family members may represent a critical regulatory checkpoint to enable microglia to achieve efficient cell cycling. Furthermore, analysis of the activity profile of the repertoire of promoter-distal genomic regulatory elements suggests a relatively restricted role for these elements in coordinating cell cycle gene expression in microglia. Overall, proliferating microglia integrates regulation of cell cycle gene expression with their broader, context-dependent, transcriptional landscape.


Subject(s)
Gene Expression Regulation , Microglia , Animals , Cell Proliferation/genetics , Mice , Microglia/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
6.
J Neurovirol ; 27(2): 279-301, 2021 04.
Article in English | MEDLINE | ID: mdl-33646495

ABSTRACT

HIV-1 infection in the central nervous system (CNS) causes the release of neurotoxic products from infected cells which trigger extensive neuronal loss. Clinically, this results in HIV-1-associated neurocognitive disorders (HAND). However, the effects on neuroprotective factors in the brain remain poorly understood and understudied in this situation. HAND is a multifactorial process involving several players, and the complex cellular mechanisms have not been fully elucidated yet. In this study, we reported that HIV-1 infection of astrocytes limits their potential to express the protective chemokine fractalkine in response to an inflammatory environment. We next confirmed that this effect was not due to a default in its shedding from the cell surface. We then investigated the biological mechanism responsible for this reduced fractalkine expression and found that HIV-1 infection specifically blocks the interaction of transcription factor NF-κB on its promoter with no effect on other cytokines. Moreover, we demonstrated that fractalkine production in astrocytes is regulated in response to immune factors secreted by infected/activated microglia and macrophages. In contrast, we observed that conditioned media from these infected cells also trigger neuronal apoptosis. At last, we demonstrated a strong neuroprotective action of fractalkine on human neurons by reducing neuronal damages. Taken together, our results indicate new relevant interactions between HIV-1 and fractalkine signaling in the CNS. This study provides new information to broaden the understanding of HAND and possibly foresee new therapeutic strategies. Considering its neuro-protective functions, reducing its production from astrocytes could have important outcomes in chronic neuroinflammation and in HIV-1 neuropathogenesis.


Subject(s)
AIDS Dementia Complex/metabolism , Astrocytes/virology , Chemokine CX3CL1/biosynthesis , Astrocytes/immunology , Astrocytes/metabolism , Cells, Cultured , HIV-1 , Humans
7.
iScience ; 24(3): 102241, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33748722

ABSTRACT

Genome-wide association studies for calcific aortic valve stenosis (CAVS) previously reported strong signal for noncoding variants at 1p21.2. Previous study using Mendelian randomization suggested that the locus controls the expression of PALMD encoding Palmdelphin (PALMD). However, the molecular regulation at the locus and the impact of PALMD on the biology of the aortic valve is presently unknown. 3D genetic mapping and CRISPR activation identified rs6702619 as being located in a distant-acting enhancer, which controls the expression of PALMD. DNA-binding assay showed that the risk variant modified the DNA shape, which prevented the recruitment of NFATC2 and lowered the expression of PALMD. In co-expression network analysis, a module encompassing PALMD was enriched in actin-based process. Mass spectrometry and functional assessment showed that PALMD is a regulator of actin polymerization. In turn, lower level of PALMD promoted the activation of myocardin-related transcription factor and fibrosis, a key pathobiological process underpinning CAVS.

8.
J Neurosci ; 41(15): 3366-3385, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33712513

ABSTRACT

Excessive inflammation within the CNS is injurious, but an immune response is also required for regeneration. Macrophages and microglia adopt different properties depending on their microenvironment, and exposure to IL4 and IL13 has been used to elicit repair. Unexpectedly, while LPS-exposed macrophages and microglia killed neural cells in culture, the addition of LPS to IL4/IL13-treated macrophages and microglia profoundly elevated IL10, repair metabolites, heparin binding epidermal growth factor trophic factor, antioxidants, and matrix-remodeling proteases. In C57BL/6 female mice, the generation of M(LPS/IL4/IL13) macrophages required TLR4 and MyD88 signaling, downstream activation of phosphatidylinositol-3 kinase/mTOR and MAP kinases, and convergence on phospho-CREB, STAT6, and NFE2. Following mouse spinal cord demyelination, local LPS/IL4/IL13 deposition markedly increased lesional phagocytic macrophages/microglia, lactate and heparin binding epidermal growth factor, matrix remodeling, oligodendrogenesis, and remyelination. Our data show that a prominent reparative state of macrophages/microglia is generated by the unexpected integration of pro- and anti-inflammatory activation cues. The results have translational potential, as the LPS/IL4/IL13 mixture could be locally applied to a focal CNS injury to enhance neural regeneration and recovery.SIGNIFICANCE STATEMENT The combination of LPS and regulatory IL4 and IL13 signaling in macrophages and microglia produces a previously unknown and particularly reparative phenotype devoid of pro-inflammatory neurotoxic features. The local administration of LPS/IL4/IL13 into spinal cord lesion elicits profound oligodendrogenesis and remyelination. The careful use of LPS and IL4/IL13 mixture could harness the known benefits of neuroinflammation to enable repair in neurologic insults.


Subject(s)
Macrophages/metabolism , Microglia/metabolism , Myelin Sheath/metabolism , Signal Transduction , Spinal Cord Regeneration , Spinal Cord/metabolism , Animals , Cells, Cultured , Coculture Techniques/methods , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Inflammation , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/toxicity , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Myeloid Differentiation Factor 88/metabolism , NF-E2 Transcription Factor, p45 Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , STAT6 Transcription Factor/metabolism , Spinal Cord/pathology , Spinal Cord/physiology , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/metabolism
9.
Cell Rep ; 33(13): 108560, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378678

ABSTRACT

The role of RNA binding proteins in regulating the phagocytic and cytokine-releasing functions of microglia is unknown. Here, we show that microglia deficient for the QUAKING (QKI) RNA binding protein have increased proinflammatory cytokine release and defects in processing phagocytosed cargo. Splicing analysis reveals a role for QKI in regulating microexon networks of the Rho GTPase pathway. We show an increase in RhoA activation and proinflammatory cytokines in QKI-deficient microglia that are repressed by treating with a Rock kinase inhibitor. During the cuprizone diet, mice with QKI-deficient microglia are inefficient at supporting central nervous system (CNS) remyelination and cause the recruited oligodendrocyte precursor cells to undergo apoptosis. Furthermore, the expression of QKI in microglia is downregulated in preactive, chronic active, and remyelinating white matter lesions of multiple sclerosis (MS) patients. Overall, our findings identify QKI as an alternative splicing regulator governing a network of Rho GTPase microexons with implications for CNS remyelination and MS patients.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Microglia/physiology , RNA-Binding Proteins/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Central Nervous System/metabolism , Cytokines/metabolism , Female , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Multiple Sclerosis/genetics , Phagocytosis , RNA/metabolism , RNA-Seq , Remyelination , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
10.
Immunity ; 52(6): 1057-1074.e7, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32362324

ABSTRACT

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macrophage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms controlling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages during diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These findings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited macrophages to acquire distinct gene expression programs and corresponding functions.


Subject(s)
Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Gene Expression Regulation , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Chromatin Immunoprecipitation Sequencing , Diet , Disease Models, Animal , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Kupffer Cells/immunology , Kupffer Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Protein Binding , Signal Transduction , Single-Cell Analysis
11.
Sci Rep ; 10(1): 6261, 2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32253373

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Glia ; 68(8): 1643-1654, 2020 08.
Article in English | MEDLINE | ID: mdl-31994799

ABSTRACT

Microglia perform multiple tasks that are essential to ensure proper cerebral functions, including synaptic remodeling, clearance of molecular debris, prevention of infections, and so forth. Furthermore, accumulating genetic and pathological evidence implicates microglial cell activity in the etiology of numerous neurodegenerative diseases and psychiatric disorders. Given this, efforts aimed at understanding the molecular mechanisms underlying microglial cell functions hold great potential for the development of novel therapies for various conditions affecting the central nervous system. In that regard, the application of paradigms in epigenomics to study transcription in microglia has provided significant insights into the molecular mechanisms that control the ontogeny and functions of these cells. With a focus on the roles of genomic regulatory elements and the epigenetic marks that control microglial gene expression, we review here recent key advancements in our comprehension of the epigenomic and transcriptional mechanisms that enable microglial cell development and activity.


Subject(s)
Central Nervous System/metabolism , Microglia/metabolism , Neurodegenerative Diseases/pathology , Neurogenesis/physiology , Animals , Brain/metabolism , Epigenomics/methods , Humans , Neurodegenerative Diseases/metabolism
13.
Life Sci Alliance ; 2(6)2019 12.
Article in English | MEDLINE | ID: mdl-31792059

ABSTRACT

CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.


Subject(s)
Aging, Premature/genetics , CX3C Chemokine Receptor 1/deficiency , Microglia/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Female , Gene Deletion , Genetic Profile , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Receptors, Chemokine/deficiency , Signal Transduction , Transcriptome
14.
Science ; 366(6469): 1134-1139, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31727856

ABSTRACT

Noncoding genetic variation is a major driver of phenotypic diversity, but functional interpretation is challenging. To better understand common genetic variation associated with brain diseases, we defined noncoding regulatory regions for major cell types of the human brain. Whereas psychiatric disorders were primarily associated with variants in transcriptional enhancers and promoters in neurons, sporadic Alzheimer's disease (AD) variants were largely confined to microglia enhancers. Interactome maps connecting disease-risk variants in cell-type-specific enhancers to promoters revealed an extended microglia gene network in AD. Deletion of a microglia-specific enhancer harboring AD-risk variants ablated BIN1 expression in microglia, but not in neurons or astrocytes. These findings revise and expand the list of genes likely to be influenced by noncoding variants in AD and suggest the probable cell types in which they function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Brain/metabolism , Enhancer Elements, Genetic/genetics , Genetic Variation , Microglia/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics , Cells, Cultured , Chromatin/metabolism , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Sequence Deletion
15.
Wiley Interdiscip Rev Syst Biol Med ; 11(5): e1449, 2019 09.
Article in English | MEDLINE | ID: mdl-31016893

ABSTRACT

Microglia are the specialized macrophages of the brain and play essential roles in ensuring its proper functioning. Accumulating evidence suggests that these cells coordinate the inflammatory response that accompanies various clinical brain conditions, including neurodegenerative diseases and psychiatric disorders. Therefore, investigating the functions of these cells and how these are regulated have become important areas of research in neuroscience over the past decade. In this regards, recent efforts to characterize the epigenomic mechanisms underlying microglial gene transcription have provided significant insights into the mechanisms that control the ontogeny and the cellular competences of microglia. In particular, these studies have established that a substantial proportion of the microglial repertoire of promoter-distal genomic regulatory elements, or enhancers, is relatively specific to these cells compared to other tissue-resident macrophages. Notably, this specificity is under the regulation of factors present in the brain that modulate activity of target axes of signaling pathways-transcription factors in microglia. Thus, the microglial enhancer repertoire is highly responsive to perturbations in the cerebral tissue microenvironment and this responsiveness has profound implications on the activity of these cells in brain diseases. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Models of Systems Properties and Processes > Mechanistic Models Biological Mechanisms > Cell Fates Developmental Biology > Lineages.


Subject(s)
Enhancer Elements, Genetic/genetics , Microglia/metabolism , Animals , Brain/metabolism , Brain Diseases/genetics , Brain Diseases/pathology , Histone Deacetylases/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Microglia/cytology , Signal Transduction/genetics , Transcription Factors/metabolism
16.
Nat Neurosci ; 22(2): 243-255, 2019 02.
Article in English | MEDLINE | ID: mdl-30617258

ABSTRACT

Autism spectrum disorder (ASD) is thought to emerge during early cortical development. However, the exact developmental stages and associated molecular networks that prime disease propensity are elusive. To profile early neurodevelopmental alterations in ASD with macrocephaly, we monitored subject-derived induced pluripotent stem cells (iPSCs) throughout the recapitulation of cortical development. Our analysis revealed ASD-associated changes in the maturational sequence of early neuron development, involving temporal dysregulation of specific gene networks and morphological growth acceleration. The observed changes tracked back to a pathologically primed stage in neural stem cells (NSCs), reflected by altered chromatin accessibility. Concerted over-representation of network factors in control NSCs was sufficient to trigger ASD-like features, and circumventing the NSC stage by direct conversion of ASD iPSCs into induced neurons abolished ASD-associated phenotypes. Our findings identify heterochronic dynamics of a gene network that, while established earlier in development, contributes to subsequent neurodevelopmental aberrations in ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Gene Regulatory Networks , Inhibitory Postsynaptic Potentials/physiology , Nerve Net/physiopathology , Neurons/physiology , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Humans , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Neurons/pathology
17.
Neuron ; 100(4): 777-779, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30465763

ABSTRACT

In a provocative study recently published in Nature, Bussian et al. (2018) report that in tauopathies, glial cells, but not neurons, may be prone to enter cell senescence. Importantly, eliminating senescent glial cells preserves neuronal functions and prevents cognitive decline.


Subject(s)
Cellular Senescence/physiology , Neurodegenerative Diseases/metabolism , Neuroglia/metabolism , Animals , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neuroglia/pathology , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology
18.
Sci Rep ; 8(1): 10757, 2018 Jul 17.
Article in English | MEDLINE | ID: mdl-30018301

ABSTRACT

Microglia are the main immune cells of the brain and express a large genetic pattern of genes linked to Parkinson's disease risk alleles. Monocytes like microglia are myeloid-lineage cells, raising the questions of the extent to which they share gene expression with microglia and whether they are already altered early in the clinical course of the disease. To decipher a monocytic gene expression signature in Parkinson's disease, we performed RNA-seq and applied the two-sample Kolmogorov-Smirnov test to identify differentially expressed genes between controls and patients with Parkinson's disease and changes in gene expression variability and dysregulation. The gene expression profiles of normal human monocytes and microglia showed a plethora of differentially expressed genes. Additionally, we identified a distinct gene expression pattern of monocytes isolated from Parkinson's disease patients at an early disease stage compared to controls using the Kolmogorov-Smirnov test. Differentially expressed genes included genes involved in immune activation such as HLA-DQB1, MYD88, REL, and TNF-α. Our data suggest that future studies of distinct leukocyte subsets are warranted to identify possible surrogate biomarkers and may lead to the identification of novel interventions early in the disease course.


Subject(s)
Monocytes/metabolism , Parkinson Disease/genetics , Biomarkers/metabolism , Gene Expression Profiling , Humans , Male , Middle Aged , Sequence Analysis, RNA , Transcriptome
19.
Anal Chem ; 89(19): 10124-10128, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28895397

ABSTRACT

Nucleic acid amplification testing is a very powerful method to perform efficient and early diagnostics. However, the integration of a DNA amplification reaction with its associated detection in a low-cost, portable, and autonomous device remains challenging. Addressing this challenge, the use of screen-printed electrochemical sensor is reported. To achieve the detection of the DNA amplification reaction, a real-time monitoring of the hydronium ions concentration, a byproduct of this reaction, is performed. Such measurements are done by potentiometry using polyaniline (PAni)-based working electrodes and silver/silver chloride reference electrodes. The developed potentiometric sensor is shown to enable the real-time monitoring of a loop-mediated isothermal amplification (LAMP) reaction with an initial number of DNA strands as low as 10 copies. In addition, the performance of this PAni-based sensor is compared to fluorescence measurements, and it is shown that similar results are obtained for both methods.


Subject(s)
Aniline Compounds/chemistry , DNA/analysis , Nucleic Acid Amplification Techniques/methods , Electrochemical Techniques , Electrodes , Hydrogen-Ion Concentration , Microfluidics
20.
Science ; 356(6344)2017 06 23.
Article in English | MEDLINE | ID: mdl-28546318

ABSTRACT

Microglia play essential roles in central nervous system (CNS) homeostasis and influence diverse aspects of neuronal function. However, the transcriptional mechanisms that specify human microglia phenotypes are largely unknown. We examined the transcriptomes and epigenetic landscapes of human microglia isolated from surgically resected brain tissue ex vivo and after transition to an in vitro environment. Transfer to a tissue culture environment resulted in rapid and extensive down-regulation of microglia-specific genes that were induced in primitive mouse macrophages after migration into the fetal brain. Substantial subsets of these genes exhibited altered expression in neurodegenerative and behavioral diseases and were associated with noncoding risk variants. These findings reveal an environment-dependent transcriptional network specifying microglia-specific programs of gene expression and facilitate efforts to understand the roles of microglia in human brain diseases.


Subject(s)
Environment , Gene Regulatory Networks/physiology , Microglia/cytology , Microglia/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Cells, Cultured , Epilepsy/genetics , Epilepsy/physiopathology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...