Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cell ; 153(3): 707-20, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23622250

RESUMEN

The genetics of complex disease produce alterations in the molecular interactions of cellular pathways whose collective effect may become clear through the organized structure of molecular networks. To characterize molecular systems associated with late-onset Alzheimer's disease (LOAD), we constructed gene-regulatory networks in 1,647 postmortem brain tissues from LOAD patients and nondemented subjects, and we demonstrate that LOAD reconfigures specific portions of the molecular interaction structure. Through an integrative network-based approach, we rank-ordered these network structures for relevance to LOAD pathology, highlighting an immune- and microglia-specific module that is dominated by genes involved in pathogen phagocytosis, contains TYROBP as a key regulator, and is upregulated in LOAD. Mouse microglia cells overexpressing intact or truncated TYROBP revealed expression changes that significantly overlapped the human brain TYROBP network. Thus the causal network structure is a useful predictor of response to gene perturbations and presents a framework to test models of disease mechanisms underlying LOAD.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Redes Reguladoras de Genes , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Teorema de Bayes , Encéfalo/patología , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Microglía/metabolismo
2.
Glia ; 69(7): 1619-1636, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33340149

RESUMEN

Sialic acids are monosaccharides that normally terminate the glycan chains of cell surface glyco-proteins and -lipids in mammals, and are highly enriched in the central nervous tissue. Sialic acids are conjugated to proteins and lipids (termed "sialylation") by specific sialyltransferases, and are removed ("desialylation") by neuraminidases. Cell surface sialic acids are sensed by complement factor H (FH) to inhibit complement activation or by sialic acid-binding immunoglobulin-like lectin (SIGLEC) receptors to inhibit microglial activation, phagocytosis, and oxidative burst. In contrast, desialylation of cells enables binding of the opsonins C1, calreticulin, galectin-3, and collectins, stimulating phagocytosis of such cells. Hypersialylation is used by bacteria and cancers as camouflage to escape immune recognition, while polysialylation of neurons protects synapses and neurogenesis. Insufficient lysosomal cleavage of sialylated molecules can lead to lysosomal accumulation of lipids and aggregated proteins, which if excessive may be expelled into the extracellular space. On the other hand, desialylation of immune receptors can activate them or trigger removal of proteins. Loss of inhibitory SIGLECs or FH triggers reduced clearance of aggregates, oxidative brain damage and complement-mediated retinal damage. Thus, cell surface sialylation recognized by FH, SIGLEC, and other immune-related receptors acts as a major checkpoint inhibitor of innate immune responses in the central nervous system, while excessive cleavage of sialic acid residues and consequently removing this checkpoint inhibitor may trigger lipid accumulation, protein aggregation, inflammation, and neurodegeneration.


Asunto(s)
Inmunidad Innata , Fagocitosis , Animales , Sistema Nervioso Central/metabolismo , Fagocitosis/fisiología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Ácidos Siálicos/metabolismo
3.
Glia ; 69(1): 137-150, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32721081

RESUMEN

Repeated systemic challenge with lipopolysaccharides (LPS) can induce microglia activation and inflammatory neurodegeneration in the substantia nigra pars compacta region of mice. We now explored the role of mononuclear phagocytes associated nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX-2) in inflammatory neurodegeneration. Cybb-deficient NOX-2 knock-out (KO) and control wild type (WT) mice were treated intraperitoneally daily over four consecutive days with 1 µg/gbw/day LPS. Transcriptome analysis by RNA-seq of total brain tissue indicated increased LPS-induced upregulation of genes belonging to the reactive oxygen species and reactive nitrogen species production, complement and lysosome activation as well as apoptosis and necroptosis in WT compared to NOX-2 KO mice. Validation of up-regulated gene transcripts via qRT-PCR confirmed that LPS-challenged NOX-2 KO mice expressed lower levels of the microglial phagocytosis-related genes Nos2, Cd68, Aif1/Iba1, Cyba, Itgam, and Fcer1g compared to WT mice at Day 5 after systemic inflammatory challenge, but no significant differences in the pro-inflammatory genes Tnfα and Il1b as well as microglial IBA1 and CD68 intensities were observed between both genotypes. Furthermore, loss of tyrosine hydroxylase positive (TH+) and NeuN positive neurons in the substantia nigra pars compacta upon repeated systemic LPS application were attenuated in NOX-2 KO mice. Thus, our data demonstrate that loss of dopaminergic neurons in the substantia nigra pars compacta after repeated systemic challenge with LPS is associated with a microglial phagocytosis-related gene activation profile involving the NADPH oxidase subunit Cybb/gp91phox.


Asunto(s)
Microglía , Fagocitosis , Animales , Neuronas Dopaminérgicas , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , NADPH Oxidasa 2/genética , NADPH Oxidasas/genética , Receptores Inmunológicos
4.
Glia ; 69(12): 2845-2862, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34406679

RESUMEN

Parkinson's disease is one of the most common neurodegenerative diseases in the elderly population, with a pathophysiology linked to neuroinflammation, complement activation, and oxidative damage. Soluble polysialic acid with an average degree of polymerization 20 (polySia avDP20) prevents inflammation and oxidative burst in human macrophages via sialic acid-binding immunoglobulin like lectin-11 (SIGLEC11) receptor and interferes with alternative complement activation. Here, we confirmed the anti-inflammatory capacity of polySia avDP20 on cultured murine embryonic stem cell-derived microglia and analyzed the effect of polySia avDP20 in a lipopolysaccharide-triggered animal model of Parkinson's disease. We demonstrated a neuroprotective effect of intraperitoneally applied polySia avDP20 in humanized SIGLEC11 transgenic mice after repeated systemic challenge with lipopolysaccharide. Pathway enrichment analysis of the brain transcriptome on day 19 after disease initiation showed that intraperitoneal application of 10 µg/g body weight polySia avDP20 prevented excessive inflammation. In line with these data, polySia avDP20 attenuated the lipopolysaccharide-triggered increase in mRNA levels of immune-related genes (Il1b, Cd14, Myd88, Fcer1g, Itgam, C4, Cybb, Iba1 and Cd68) and cell death-related genes (Casp8, Ripk1 and Ripk3) in the brains of SIGLEC11 transgenic mice on day 19, but not on day 5. Moreover, immunohistochemistry demonstrated that polySia avDP20 reduced the lipopolysaccharide-induced increase in immunoreactivity of IBA1 and CD68 in the substantia nigra pars reticulata in SIGLEC11 transgenic and wild type mice on day 19. Furthermore, treatment with polySia avDP20 prevented the loss of dopaminergic neurons in the substantia nigra pars compacta induced by lipopolysaccharide challenge in both SIGLEC11 transgenic and wild type mice on day 19. Thus, our data demonstrate that polySia avDP20 ameliorates inflammatory dopaminergic neurodegeneration and therefore is a promising drug candidate to prevent Parkinson's disease-related inflammation and neurodegeneration.


Asunto(s)
Lipopolisacáridos , Ácidos Siálicos , Anciano , Animales , Neuronas Dopaminérgicas , Humanos , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Peso Molecular , Ácidos Siálicos/farmacología
5.
Glia ; 69(6): 1393-1412, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33539598

RESUMEN

Genome-wide association studies demonstrated that polymorphisms in the CD33/sialic acid-binding immunoglobulin-like lectin 3 gene are associated with late-onset Alzheimer's disease (AD). CD33 is expressed on myeloid immune cells and mediates inhibitory signaling through protein tyrosine phosphatases, but the exact function of CD33 in microglia is still unknown. Here, we analyzed CD33 knockout human THP1 macrophages and human induced pluripotent stem cell-derived microglia for immunoreceptor tyrosine-based activation motif pathway activation, cytokine transcription, phagocytosis, and phagocytosis-associated oxidative burst. Transcriptome analysis of the macrophage lines showed that knockout of CD33 as well as knockdown of the CD33 signaling-associated protein tyrosine phosphatase, nonreceptor type 6 (PTPN6) led to constitutive activation of inflammation-related pathways. Moreover, deletion of CD33 or expression of Exon 2-deleted CD33 (CD33ΔE2 /CD33m) led to increased phosphorylation of the kinases spleen tyrosine kinase (SYK) and extracellular signal-regulated kinase 1 and 2 (ERK1 and 2). Transcript analysis by quantitative real-time polymerase chain reaction confirmed increased levels of interleukin (IL) 1B, IL8, and IL10 after knockout of CD33 in macrophages and microglia. In addition, upregulation of the gene transcripts of the AD-associated phosphatase INPP5D was observed after knockout of CD33. Functional analysis of macrophages and microglia showed that phagocytosis of aggregated amyloid-ß1-42 and bacterial particles were increased after knockout of CD33 or CD33ΔE2 expression and knockdown of PTPN6. Furthermore, the phagocytic oxidative burst during uptake of amyloid-ß1-42 or bacterial particles was increased after CD33 knockout but not in CD33ΔE2 -expressing microglia. In summary, deletion of CD33 or expression of CD33ΔE2 in human macrophages and microglia resulted in putative beneficial phagocytosis of amyloid ß1-42 , but potentially detrimental oxidative burst and inflammation, which was absent in CD33ΔE2 -expressing microglia.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Estudio de Asociación del Genoma Completo , Humanos , Inflamación , Microglía , Fenotipo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética
6.
Glia ; 69(5): 1126-1139, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33314333

RESUMEN

The triggering receptor expressed on myeloid cells 2 (TREM2) is an immune receptor expressed on myeloid-derived cell types. The extracellular immunoglobulin-like domain of TREM2 binds anionic ligands including Apolipoprotein E and Amyloid-ß. The transmembrane domain interacts with its adaptor protein DAP12/TYROBP that is responsible for propagation of downstream signaling upon ligand interaction. Several sequence variants of TREM2 have been linked to different neurodegenerative diseases including Alzheimer's disease. Here, we generated HEK 293 Flp-In cell lines stably expressing human TREM2 and DAP12 using a bicistronic construct with a T2A linker sequence allowing initial expression of both proteins in stoichiometric amounts. Cell biological and biochemical analyses revealed transport of TREM2 to the cell surface, and canonical sequential proteolytic processing and shedding of TREM2 (sTREM2). The functionality of this cell system was demonstrated by detection of phosphorylated spleen tyrosine kinase (SYK) upon stimulation of TREM2 with the anionic membrane lipid phosphatidylserine or anti-TREM2 antibodies. Using this cell model, we demonstrated impaired signaling of disease associated TREM2 variants. We also identified a monoclonal antibody against the stalk region of TREM2 with agonistic activity. Activation of TREM2-DAP12 signaling with the monoclonal antibody and the partial loss of function of disease associated variants were recapitulated in induced pluripotent stem cell derived microglia. Thus, this reporter cell model represents a suitable experimental system to investigate signaling of TREM2 variants, and for the identification of ligands and compounds that modulate TREM2-DAP12 signaling. MAIN POINTS: Disease associated variants impair the signaling activity of TREM2 by distinct mechanisms. Targeting the stalk region of TREM2 with bivalent antibodies activates TREM2 signaling.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Anticuerpos Monoclonales , Proteínas Portadoras , Células HEK293 , Humanos , Ligandos , Glicoproteínas de Membrana/genética , Células Mieloides , Receptores Inmunológicos/genética
7.
Glycobiology ; 31(11): 1478-1489, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34224569

RESUMEN

Among the enzymes of the biosynthesis of sialoglycoconjugates, uridine diphosphate-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE), catalyzing the first essential step of the sialic acid (Sia) de novo biosynthesis, and cytidine monophosphate (CMP)-Sia synthase (CMAS), activating Sia to CMP-Sia, are particularly important. The knockout of either of these enzymes in mice is embryonically lethal. While the lethality of Cmas-/- mice has been attributed to a maternal complement attack against asialo fetal placental cells, the cause of lethality in Gne-deficient embryos has remained elusive. Here, we advanced the significance of sialylation for embryonic development through detailed histological analyses of Gne-/- embryos and placentae. We found that Gne-/- embryonic and extraembryonic tissues are hyposialylated rather than being completely deficient of sialoglycans, which holds true for Cmas-/- embryos. Residual sialylation of Gne-/- cells can be explained by scavenging free Sia from sialylated maternal serum glycoconjugates via the lysosomal salvage pathway. The placental architecture of Gne-/- mice was unaffected, but severe hemorrhages in the neuroepithelium with extensive bleeding into the cephalic ventricles were present at E12.5 in the mutants. At E13.5, the vast majority of Gne-/- embryos were asystolic. This phenotype persisted when Gne-/- mice were backcrossed to a complement component 3-deficient background, confirming distinct pathomechanisms of Cmas-/- and Gne-/- mice. We conclude that the low level of sialylation observed in Gne-/- mice is sufficient both for immune homeostasis at the fetal-maternal interface and for embryonic development until E12.5. However, formation of the neural microvasculature is the first critical process, depending on a higher degree of sialylation during development of the embryo proper.


Asunto(s)
Hemorragia Cerebral/metabolismo , Complejos Multienzimáticos/metabolismo , Animales , Biocatálisis , Hemorragia Cerebral/patología , Desarrollo Embrionario , Ratones , Ratones Noqueados , Complejos Multienzimáticos/deficiencia , Ácido N-Acetilneuramínico/biosíntesis
8.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32752058

RESUMEN

Sialic acids (Sias) are the most abundant terminal sugar residues of glycoproteins and glycolipids on the surface of mammalian cells. The nervous tissue is the organ with the highest expression level of Sias. The 'sialylation' of glycoconjugates is performed via sialyltransferases, whereas 'desialylation' is done by sialidases or is a possible consequence of oxidative damage. Sialic acid residues on the neural cell surfaces inhibit complement and microglial activation, as well as phagocytosis of the underlying structures, via binding to (i) complement factor H (CFH) or (ii) sialic acid-binding immunoglobulin-like lectin (SIGLEC) receptors. In contrast, activated microglial cells show sialidase activity that desialylates both microglia and neurons, and further stimulates innate immunity via microglia and complement activation. The desialylation conveys neurons to become susceptible to phagocytosis, as well as triggers a microglial phagocytosis-associated oxidative burst and inflammation. Dysfunctions of the 'Sia-SIGLEC' and/or 'Sia-complement' axes often lead to neurological diseases. Thus, Sias on glycoconjugates of the intact glycocalyx and its desialylation are major regulators of neuroinflammation.


Asunto(s)
Inmunidad Innata/genética , Tejido Nervioso/metabolismo , Ácidos Siálicos/genética , Sialiltransferasas/genética , Glicoconjugados/genética , Glicoconjugados/inmunología , Humanos , Macrófagos , Microglía/inmunología , Microglía/metabolismo , Tejido Nervioso/inmunología , Neuronas/metabolismo , Neuronas/patología , Fagocitosis/genética , Ácidos Siálicos/inmunología , Ácidos Siálicos/metabolismo , Sialiltransferasas/inmunología
9.
Glia ; 67(3): 539-550, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30548312

RESUMEN

The microglial triggering receptor expressed on myeloid cells 2 (TREM2) signals via the activatory membrane adaptor molecule TYROBP. Genetic variants or mutations of TREM2 or TYROBP have been linked to inflammatory neurodegenerative diseases associated with aging. The typical aging process goes along with microglial changes and mild neuronal loss, but the exact contribution of TREM2 is still unclear. Aged TREM2 knock-out mice showed decreased age-related neuronal loss in the substantia nigra and the hippocampus. Transcriptomic analysis of the brains of 24 months old TREM2 knock-out mice revealed 211 differentially expressed genes mostly downregulated and associated with complement activation and oxidative stress response pathways. Consistently, 24 months old TREM2 knock-out mice showed lower transcription of microglial (Aif1 and Tmem119), oxidative stress markers (Inos, Cyba, and Cybb) and complement components (C1qa, C1qb, C1qc, C3, C4b, Itgam, and Itgb2), decreased microglial numbers and expression of the microglial activation marker Cd68, as well as accumulation of oxidized lipids. Cultured microglia of TREM2 knock-out mice showed reduced phagocytosis and oxidative burst. Thus, microglial TREM2 contributes to age-related microglial changes, phagocytic oxidative burst, and loss of neurons with possible detrimental effects during physiological aging.


Asunto(s)
Envejecimiento/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptores Inmunológicos/metabolismo , Factores de Edad , Envejecimiento/genética , Animales , Hipocampo/citología , Hipocampo/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Microglía/citología , Neuronas/citología , Estrés Oxidativo/fisiología , Fagocitosis/fisiología , Receptores Inmunológicos/genética , Sustancia Negra/citología , Sustancia Negra/metabolismo
10.
Glia ; 66(10): 2246-2261, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30277599

RESUMEN

Chemokines are important signaling molecules in the immune and nervous system. Using a fluorescence reporter mouse model, we demonstrate that the chemokine CCL17, a ligand of the chemokine receptor CCR4, is produced in the murine brain, particularly in a subset of hippocampal CA1 neurons. We found that basal expression of Ccl17 in hippocampal neurons was strongly enhanced by peripheral challenge with lipopolysaccharide (LPS). LPS-mediated induction of Ccl17 in the hippocampus was dependent on local tumor necrosis factor (TNF) signaling, whereas upregulation of Ccl22 required granulocyte-macrophage colony-stimulating factor (GM-CSF). CCL17 deficiency resulted in a diminished microglia density under homeostatic and inflammatory conditions. Further, microglia from naïve Ccl17-deficient mice possessed a reduced cellular volume and a more polarized process tree as assessed by computer-assisted imaging analysis. Regarding the overall branching, cell surface area, and total tree length, the morphology of microglia from naïve Ccl17-deficient mice resembled that of microglia from wild-type mice after LPS stimulation. In line, electrophysiological recordings indicated that CCL17 downmodulates basal synaptic transmission at CA3-CA1 Schaffer collaterals in acute slices from naïve but not LPS-treated animals. Taken together, our data identify CCL17 as a homeostatic and inducible neuromodulatory chemokine affecting the presence and morphology of microglia and synaptic transmission in the hippocampus.


Asunto(s)
Quimiocina CCL17/metabolismo , Hipocampo/inmunología , Neuroinmunomodulación/fisiología , Neuronas/inmunología , Animales , Quimiocina CCL17/genética , Quimiocina CCL22/metabolismo , Femenino , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/patología , Homeostasis/fisiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Monocitos/inmunología , Monocitos/patología , Neuronas/patología , Receptores CCR4/metabolismo , Transmisión Sináptica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Glia ; 65(7): 1103-1118, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28370426

RESUMEN

The Eph-ephrin system plays pivotal roles in cell adhesion and migration. The receptor-like functions of the ephrin ligands allow the regulation of intracellular processes via reverse signaling. γ-Secretase mediated processing of ephrin-B has previously been linked to activation of Src, a kinase crucial for focal adhesion and podosome phosphorylation. Here, we analyzed the role of γ-secretase in the stimulation of reverse ephrin-B2 signaling in the migration of mouse embryonic stem cell derived microglia. The proteolytic generation of the ephrin-B2 intracellular domain (ICD) by γ-secretase stimulates Src and focal adhesion kinase (FAK). Inhibition of γ-secretase decreased the phosphorylation of Src and FAK, and reduced cell motility. These effects were associated with enlargement of the podosomal surface. Interestingly, expression of ephrin-B2 ICD could rescue these effects, indicating that this proteolytic fragment mediates the activation of Src and FAK, and thereby regulates podosomal dynamics in microglial cells. Together, these results identify γ-secretase as well as ephrin-B2 as regulators of microglial migration.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Movimiento Celular/fisiología , Citoplasma/metabolismo , Efrina-B2/metabolismo , Microglía/citología , Microglía/fisiología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Animales Recién Nacidos , Movimiento Celular/genética , Embrión de Mamíferos , Efrina-B2/genética , Quinasa 1 de Adhesión Focal/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Fosforilación , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor EphB1/metabolismo , Transducción de Señal/genética , Células Madre/fisiología
12.
J Neuroinflammation ; 14(1): 4, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28086806

RESUMEN

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. The polymorphism rs10490924 in the ARMS2 gene is highly associated with AMD and linked to an indel mutation (del443ins54), the latter inducing mRNA instability. At present, the function of the ARMS2 protein, the exact cellular sources in the retina and the biological consequences of the rs10490924 polymorphism are unclear. METHODS: Recombinant ARMS2 was expressed in Pichia pastoris, and protein functions were studied regarding cell surface binding and complement activation in human serum using fluoresence-activated cell sorting (FACS) as well as laser scanning microscopy (LSM). Biolayer interferometry defined protein interactions. Furthermore, endogenous ARMS2 gene expression was studied in human blood derived monocytes and in human induced pluripotent stem cell-derived microglia (iPSdM) by PCR and LSM. The ARMS2 protein was localized in human genotyped retinal sections and in purified monocytes derived from AMD patients without the ARMS2 risk variant by LSM. ARMS2 expression in monocytes under oxidative stress was determined by Western blot analysis. RESULTS: Here, we demonstrate for the first time that ARMS2 functions as surface complement regulator. Recombinant ARMS2 binds to human apoptotic and necrotic cells and initiates complement activation by recruiting the complement activator properdin. ARMS2-properdin complexes augment C3b surface opsonization for phagocytosis. We also demonstrate for the first time expression of ARMS2 in human monocytes especially under oxidative stress and in microglia cells of the human retina. The ARMS2 protein is absent in monocytes and also in microglia cells, derived from patients homozygous for the ARMS2 AMD risk variant (rs10490924). CONCLUSIONS: ARMS2 is likely involved in complement-mediated clearance of cellular debris. As AMD patients present with accumulated proteins and lipids on Bruch's membrane, ARMS2 protein deficiency due to the genetic risk variant might be involved in drusen formation.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Degeneración Macular/genética , Degeneración Macular/metabolismo , Polimorfismo de Nucleótido Simple/genética , Proteínas/genética , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Células CHO , Proteínas del Sistema Complemento/genética , Cricetulus , Femenino , Heparitina Sulfato/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Factores Inmunológicos/farmacología , Degeneración Macular/patología , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Properdina/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteínas/inmunología , Proteínas/metabolismo , Retina/metabolismo , Retina/patología , Adulto Joven
13.
J Immunol ; 194(7): 3400-13, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25725102

RESUMEN

A broad spectrum of diseases is characterized by myelin abnormalities and/or oligodendrocyte pathology. In most, if not all, of these diseases, early activation of microglia occurs. Our knowledge regarding the factors triggering early microglia activation is, however, incomplete. In this study, we used the cuprizone model to investigate the temporal and causal relationship of oligodendrocyte apoptosis and early microglia activation. Genome-wide gene expression studies revealed the induction of distinct chemokines, among them Cxcl10, Ccl2, and Ccl3 in cuprizone-mediated oligodendrocyte apoptosis. Early microglia activation was unchanged in CCL2- and CCL3-deficient knockouts, but was significantly reduced in CXCL10-deficient mice, resulting in an amelioration of cuprizone toxicity at later time points. Subsequent in vitro experiments revealed that recombinant CXCL10 induced migration and a proinflammatory phenotype in cultured microglia, without affecting their phagocytic activity or proliferation. In situ hybridization analyses suggest that Cxcl10 mRNA is mainly expressed by astrocytes, but also oligodendrocytes, in short-term cuprizone-exposed mice. Our results show that CXCL10 actively participates in the initiation of microglial activation. These findings have implications for the role of CXCL10 as an important mediator during the initiation of neuroinflammatory processes associated with oligodendrocyte pathology.


Asunto(s)
Quimiocina CXCL10/genética , Cuprizona/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Animales , Astrocitos/metabolismo , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocinas/genética , Quimiocinas/metabolismo , Cuprizona/administración & dosificación , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Inmunohistoquímica , Lactato Deshidrogenasas/metabolismo , Ratones , Ratones Noqueados , Microglía/inmunología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Ratas
14.
Brain ; 139(Pt 2): 338-45, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26685157

RESUMEN

Isolated cytochrome c oxidase (complex IV) deficiency is one of the most frequent respiratory chain defects in humans and is usually caused by mutations in proteins required for assembly of the complex. Mutations in nuclear-encoded structural subunits are very rare. In a patient with Leigh-like syndrome presenting with leukodystrophy and severe epilepsy, we identified a homozygous splice site mutation in COX8A, which codes for the ubiquitously expressed isoform of subunit VIII, the smallest nuclear-encoded subunit of complex IV. The mutation, affecting the last nucleotide of intron 1, leads to aberrant splicing, a frame-shift in the highly conserved exon 2, and decreased amount of the COX8A transcript. The loss of the wild-type COX8A protein severely impairs the stability of the entire cytochrome c oxidase enzyme complex and manifests in isolated complex IV deficiency in skeletal muscle and fibroblasts, similar to the frequent c.845_846delCT mutation in the assembly factor SURF1 gene. Stability and activity of complex IV could be rescued in the patient's fibroblasts by lentiviral expression of wild-type COX8A. Our findings demonstrate that COX8A is indispensable for function of human complex IV and its mutation causes human disease.


Asunto(s)
Complejo IV de Transporte de Electrones/genética , Epilepsia/diagnóstico , Epilepsia/genética , Enfermedad de Leigh/diagnóstico , Enfermedad de Leigh/genética , Subunidades de Proteína/genética , Niño , Complejo IV de Transporte de Electrones/fisiología , Epilepsia/complicaciones , Resultado Fatal , Femenino , Humanos , Enfermedad de Leigh/complicaciones , Mutación/genética
15.
Glia ; 64(1): 35-47, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26257016

RESUMEN

The complement system has been implicated in the removal of dysfunctional synapses and neurites during development and in disease processes in the mouse, but it is unclear how far the mouse data can be transferred to humans. Here, we co-cultured macrophages derived from human THP1 monocytes and neurons derived from human induced pluripotent stem cells, to study the role of the complement system in a human model. Components of the complement system were expressed by the human macrophages and human neuronal culture, while receptors of the complement cascade were expressed by human macrophages as shown via gene transcript analysis and flow cytometry. We mimicked pathological conditions leading to an altered glycocalyx by treatment of human neurons with sialidases. Desialylated human neurites were opsonized by the complement component C1q. Furthermore, human neurites with an intact sialic acid cap remained untouched, while desialylated human neurites were removed and ingested by human macrophages. While blockage of the complement receptor 1 (CD35) had no effect, blockage of CD11b as part of the complement receptor 3 (CR3) reversed the effect on macrophage phagocytosis of desialylated human neurites. Data demonstrate that in the human system sialylation of the neuronal glycocalyx serves as an inhibitory flag for complement binding and CR3-mediated phagocytosis by macrophages.


Asunto(s)
Macrófagos/fisiología , Mucolipidosis/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuritas/fisiología , Neuronas/fisiología , Fagocitosis/fisiología , Antígeno CD11b/metabolismo , Técnicas de Cocultivo , Complemento C1q/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Antígeno de Macrófago-1/metabolismo , Monocitos/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Receptores de Complemento 3b/metabolismo
16.
Glia ; 64(6): 1007-20, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26959607

RESUMEN

Microglia, the immune cells of the CNS, are highly adaptive cells that can acquire different pro- and anti-inflammatory activation states with distinct functions in CNS homeostasis and pathologies. To study microglial function in vitro, primary microglia or immortalized cell lines are commonly used. An alternative to these cells are embryonic stem cell-derived microglia (ESdM). ESdM have previously been shown to be very similar to primary microglia in terms of expression profiles and surface molecules. In this study, ESdM and primary microglia were treated with different inflammatory stimulants to analyze their ability to adopt different activation states. Using quantitative real-time PCR, comparative transcriptomics, ELISA, and flow cytometry, we found that different activation states can be induced in ESdM, which are similar to those found in primary microglia. These states are characterized by specific sets of inflammatory marker molecules and differential transcriptome signatures. Our results show that ESdM are a valuable alternative cell model to study microglial functions and neuroinflammatory mechanisms.


Asunto(s)
Microglía/citología , Células Madre Embrionarias de Ratones/citología , Transcriptoma/fisiología , Animales , Antiinflamatorios/farmacología , Biomarcadores/metabolismo , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
17.
Glia ; 64(8): 1314-30, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27159043

RESUMEN

Microglia are tissue macrophages and mediators of innate immune responses in the brain. The protein-modifying glycan polysialic acid (polySia) is implicated in modulating microglia activity. Cultured murine microglia maintain a pool of Golgi-confined polySia, which is depleted in response to lipopolysaccharide (LPS)-induced activation. Polysialylated neuropilin-2 (polySia-NRP2) contributes to this pool but further polySia protein carriers have remained elusive. Here, we use organotypic brain slice cultures to demonstrate that injury-induced activation of microglia initiates Golgi-confined polySia expression in situ. An unbiased glycoproteomic approach with stem cell-derived microglia identifies E-selectin ligand-1 (ESL-1) as a novel polySia acceptor. Together with polySia-NRP2, polySia-ESL-1 is also detected in primary cultured microglia, in brain slice cultures and in phorbol ester-induced THP-1 macrophages. Induction of stem cell-derived microglia, activated microglia in brain slice cultures and THP-1 macrophages by LPS, but not interleukin-4, causes polySia depletion and, as shown for stem cell-derived microglia, a metalloproteinase-dependent release of polySia-ESL-1 and polySia-NRP2. Moreover, soluble polySia attenuates LPS-induced production of nitric oxide and proinflammatory cytokines. Thus, shedding of polySia-ESL-1 and polySia-NRP2 after LPS-induced activation of microglia and THP-1 macrophages may constitute a mechanism for negative feedback regulation. GLIA 2016 GLIA 2016;64:1314-1330.


Asunto(s)
Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Microglía/inmunología , Neuropilina-2/metabolismo , Polisacáridos/toxicidad , Células THP-1/inmunología , Animales , Encéfalo/inmunología , Encéfalo/patología , Células Cultivadas , Citocinas/metabolismo , Escherichia coli , Humanos , Macrófagos/patología , Ratones Noqueados , Microglía/patología , Moléculas de Adhesión de Célula Nerviosa/deficiencia , Moléculas de Adhesión de Célula Nerviosa/genética , Células-Madre Neurales/inmunología , Células-Madre Neurales/patología , Óxido Nítrico/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Sialoglicoproteínas/metabolismo , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Células THP-1/patología , Técnicas de Cultivo de Tejidos
18.
J Neurosci ; 34(25): 8546-56, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24948809

RESUMEN

Systemic inflammatory reactions have been postulated to exacerbate neurodegenerative diseases via microglial activation. We now demonstrate in vivo that repeated systemic challenge of mice over four consecutive days with bacterial LPS maintained an elevated microglial inflammatory phenotype and induced loss of dopaminergic neurons in the substantia nigra. The same total cumulative LPS dose given within a single application did not induce neurodegeneration. Whole-genome transcriptome analysis of the brain demonstrated that repeated systemic LPS application induced an activation pattern involving the classical complement system and its associated phagosome pathway. Loss of dopaminergic neurons induced by repeated systemic LPS application was rescued in complement C3-deficient mice, confirming the involvement of the complement system in neurodegeneration. Our data demonstrate that a phagosomal inflammatory response of microglia is leading to complement-mediated loss of dopaminergic neurons.


Asunto(s)
Activación de Complemento/fisiología , Complemento C3/metabolismo , Proteínas del Sistema Complemento/fisiología , Neuronas Dopaminérgicas/metabolismo , Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Fagosomas/fisiología , Animales , Neuronas Dopaminérgicas/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Degeneración Nerviosa/patología , Vías Nerviosas/fisiología , Fagosomas/metabolismo , Fagosomas/patología
19.
J Neuroinflammation ; 12: 77, 2015 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-25928566

RESUMEN

BACKGROUND: Microglia reactivity is a hallmark of neurodegenerative diseases. We have previously identified activated microglia/macrophage whey acidic protein (AMWAP) as a counter-regulator of pro-inflammatory response. Here, we studied its mechanisms of action with a focus on toll-like receptor (TLR) and nuclear factor κB (NFκB) signaling. METHODS: Recombinant AMWAP was produced in Escherichia coli and HEK293 EBNA cells and purified by affinity chromatography. AMWAP uptake was identified by fluorescent labeling, and pro-inflammatory microglia markers were measured by qRT-PCR after stimulation with TLR ligands. NFκB pathway proteins were assessed by immunocytochemistry, Western blot, and immunoprecipitation. A 20S proteasome activity assay was used to investigate the anti-peptidase activity of AMWAP. Microglial neurotoxicity was estimated by nitrite measurement and quantification of caspase 3/7 levels in 661W photoreceptors cultured in the presence of microglia-conditioned medium. Microglial proliferation was investigated using flow cytometry, and their phagocytosis was monitored by the uptake of 661W photoreceptor debris. RESULTS: AMWAP was secreted from lipopolysaccharide (LPS)-activated microglia and recombinant AMWAP reduced gene transcription of IL6, iNOS, CCL2, CASP11, and TNFα in BV-2 microglia treated with LPS as TLR4 ligand. This effect was replicated with murine embryonic stem cell-derived microglia (ESdM) and primary brain microglia. AMWAP also diminished pro-inflammatory markers in microglia activated with the TLR2 ligand zymosan but had no effects on IL6, iNOS, and CCL2 transcription in cells treated with CpG oligodeoxynucleotides as TLR9 ligand. Microglial uptake of AMWAP effectively inhibited TLR4-dependent NFκB activation by preventing IRAK-1 and IκBα proteolysis. No inhibition of IκBα phosphorylation or ubiquitination and no influence on overall 20S proteasome activity were observed. Functionally, both microglial nitric oxide (NO) secretion and 661W photoreceptor apoptosis were significantly reduced after AMWAP treatment. AMWAP promoted the filopodia formation of microglia and increased the phagocytic uptake of apoptotic 661W photoreceptor cells. CONCLUSIONS: AMWAP is secreted from reactive microglia and acts in a paracrine fashion to counter-balance TLR2/TLR4-induced reactivity through NFκB inhibition. AMWAP also induces a neuroprotective microglial phenotype with reduced neurotoxicity and increased phagocytosis. We therefore hypothesize that anti-inflammatory whey acidic proteins could have a therapeutic potential in neurodegenerative diseases of the brain and the retina.


Asunto(s)
Microglía/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Leupeptinas/farmacología , Lipopolisacáridos/farmacología , Ratones , Proteínas del Tejido Nervioso/inmunología , Nitritos/metabolismo , Fagocitosis/efectos de los fármacos , Células Fotorreceptoras/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
20.
J Neurosci ; 33(46): 18270-6, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227736

RESUMEN

Sialic acid-binding Ig-like lectins (Siglecs) are members of the Ig superfamily that recognize sialic acid residues of glycoproteins. Siglec-E is a mouse CD33-related Siglec that preferentially binds to sialic acid residues of the cellular glycocalyx. Here, we demonstrate gene transcription and protein expression of Siglec-E by cultured mouse microglia. Siglec-E on microglia inhibited phagocytosis of neural debris and prevented the production of superoxide radicals induced by challenge with neural debris. Soluble extracellular Siglec-E receptor protein bound to the neural glycocalyx. Coculture of mouse microglia and neurons demonstrated a neuroprotective effect of microglial Siglec-E that was dependent on neuronal sialic acid residues. Increased neurotoxicity of microglia after knockdown of Siglece mRNA was neutralized by the reactive oxygen species scavenger Trolox. Data suggest that Siglec-E recognizes the intact neuronal glycocalyx and has neuroprotective function by preventing phagocytosis and the associated oxidative burst.


Asunto(s)
Microglía/metabolismo , Neuronas/metabolismo , Fagocitosis/fisiología , Estallido Respiratorio/fisiología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/biosíntesis , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos B/biosíntesis , Células Cultivadas , Técnicas de Cocultivo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/metabolismo , Unión Proteica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA