Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 156(4): 717-29, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24507937

RESUMEN

Acid-sensing ion channels (ASICs) detect extracellular protons produced during inflammation or ischemic injury and belong to the superfamily of degenerin/epithelial sodium channels. Here, we determine the cocrystal structure of chicken ASIC1a with MitTx, a pain-inducing toxin from the Texas coral snake, to define the structure of the open state of ASIC1a. In the MitTx-bound open state and in the previously determined low-pH desensitized state, TM2 is a discontinuous α helix in which the Gly-Ala-Ser selectivity filter adopts an extended, belt-like conformation, swapping the cytoplasmic one-third of TM2 with an adjacent subunit. Gly 443 residues of the selectivity filter provide a ring of three carbonyl oxygen atoms with a radius of ∼3.6 Å, presenting an energetic barrier for hydrated ions. The ASIC1a-MitTx complex illuminates the mechanism of MitTx action, defines the structure of the selectivity filter of voltage-independent, sodium-selective ion channels, and captures the open state of an ASIC.


Asunto(s)
Canales Iónicos Sensibles al Ácido/química , Proteínas Aviares/química , Pollos , Venenos Elapídicos/química , Elapidae , Canales Iónicos Sensibles al Ácido/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Aviares/metabolismo , Cristalografía por Rayos X , Venenos Elapídicos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia , Canales de Sodio/química
2.
Annu Rev Genet ; 53: 263-288, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31518519

RESUMEN

Advances in human genetics have implicated a growing number of genes in neurodegenerative diseases, providing insight into pathological processes. For Alzheimer disease in particular, genome-wide association studies and gene expression studies have emphasized the pathogenic contributions from microglial cells and motivated studies of microglial function/dysfunction. Here, we summarize recent genetic evidence for microglial involvement in neurodegenerative disease with a focus on Alzheimer disease, for which the evidence is most compelling. To provide context for these genetic discoveries, we discuss how microglia influence brain development and homeostasis, how microglial characteristics change in disease, and which microglial activities likely influence the course of neurodegeneration. In all, we aim to synthesize varied aspects of microglial biology and highlight microglia as possible targets for therapeutic interventions in neurodegenerative disease.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Microglía/patología , Microglía/fisiología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/fisiología , Sistema Nervioso Central/metabolismo , Vía Clásica del Complemento/fisiología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Homeostasis , Humanos , Macrófagos/fisiología , Placa Amiloide/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo
3.
J Neurosci ; 44(29)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38830764

RESUMEN

Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-ß (Aß) pathology (PS2APP) or combined Aß and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.


Asunto(s)
Enfermedad de Alzheimer , Glicoproteínas de Membrana , Microglía , Esclerosis Múltiple , Receptores Inmunológicos , Animales , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/agonistas , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Femenino , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Anticuerpos/farmacología , Humanos , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo
4.
Cell ; 141(5): 834-45, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20510930

RESUMEN

Toxins have evolved to target regions of membrane ion channels that underlie ligand binding, gating, or ion permeation, and have thus served as invaluable tools for probing channel structure and function. Here, we describe a peptide toxin from the Earth Tiger tarantula that selectively and irreversibly activates the capsaicin- and heat-sensitive channel, TRPV1. This high-avidity interaction derives from a unique tandem repeat structure of the toxin that endows it with an antibody-like bivalency. The "double-knot" toxin traps TRPV1 in the open state by interacting with residues in the presumptive pore-forming region of the channel, highlighting the importance of conformational changes in the outer pore region of TRP channels during activation.


Asunto(s)
Venenos de Araña/metabolismo , Canales Catiónicos TRPV/metabolismo , Proteínas de Xenopus/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Fenómenos Electrofisiológicos , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Neuronas/metabolismo , Oocitos/metabolismo , Ratas , Venenos de Araña/química , Canales Catiónicos TRPV/química , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Proteínas de Xenopus/química
5.
Nature ; 541(7638): 481-487, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28099414

RESUMEN

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Asunto(s)
Astrocitos/clasificación , Astrocitos/patología , Muerte Celular , Sistema Nervioso Central/patología , Microglía/patología , Neuronas/patología , Animales , Astrocitos/metabolismo , Axotomía , Técnicas de Cultivo de Célula , Supervivencia Celular , Complemento C1q/metabolismo , Progresión de la Enfermedad , Humanos , Inflamación/patología , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Enfermedades Neurodegenerativas/patología , Oligodendroglía/patología , Fagocitosis , Fenotipo , Ratas , Ratas Sprague-Dawley , Sinapsis/patología , Toxinas Biológicas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(12): E1738-46, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26884166

RESUMEN

The specific function of microglia, the tissue resident macrophages of the brain and spinal cord, has been difficult to ascertain because of a lack of tools to distinguish microglia from other immune cells, thereby limiting specific immunostaining, purification, and manipulation. Because of their unique developmental origins and predicted functions, the distinction of microglia from other myeloid cells is critically important for understanding brain development and disease; better tools would greatly facilitate studies of microglia function in the developing, adult, and injured CNS. Here, we identify transmembrane protein 119 (Tmem119), a cell-surface protein of unknown function, as a highly expressed microglia-specific marker in both mouse and human. We developed monoclonal antibodies to its intracellular and extracellular domains that enable the immunostaining of microglia in histological sections in healthy and diseased brains, as well as isolation of pure nonactivated microglia by FACS. Using our antibodies, we provide, to our knowledge, the first RNAseq profiles of highly pure mouse microglia during development and after an immune challenge. We used these to demonstrate that mouse microglia mature by the second postnatal week and to predict novel microglial functions. Together, we anticipate these resources will be valuable for the future study and understanding of microglia in health and disease.


Asunto(s)
Encéfalo/citología , Proteínas de la Membrana/análisis , Microglía/química , Proteínas del Tejido Nervioso/análisis , Anciano , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , División Celular , Linaje de la Célula , Niño , Endotoxemia/patología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Lipopolisacáridos/toxicidad , Macrófagos/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Microglía/fisiología , Persona de Mediana Edad , Compresión Nerviosa , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Traumatismos del Nervio Óptico/patología , Especificidad de Órganos , Conejos , Nervio Ciático/lesiones , Nervio Ciático/patología , Análisis de Secuencia de ARN , Lóbulo Temporal/metabolismo , Transcriptoma
7.
Nature ; 479(7373): 410-4, 2011 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22094702

RESUMEN

Natural products that elicit discomfort or pain represent invaluable tools for probing molecular mechanisms underlying pain sensation. Plant-derived irritants have predominated in this regard, but animal venoms have also evolved to avert predators by targeting neurons and receptors whose activation produces noxious sensations. As such, venoms provide a rich and varied source of small molecule and protein pharmacophores that can be exploited to characterize and manipulate key components of the pain-signalling pathway. With this in mind, here we perform an unbiased in vitro screen to identify snake venoms capable of activating somatosensory neurons. Venom from the Texas coral snake (Micrurus tener tener), whose bite produces intense and unremitting pain, excites a large cohort of sensory neurons. The purified active species (MitTx) consists of a heteromeric complex between Kunitz- and phospholipase-A2-like proteins that together function as a potent, persistent and selective agonist for acid-sensing ion channels (ASICs), showing equal or greater efficacy compared with acidic pH. MitTx is highly selective for the ASIC1 subtype at neutral pH; under more acidic conditions (pH < 6.5), MitTx massively potentiates (>100-fold) proton-evoked activation of ASIC2a channels. These observations raise the possibility that ASIC channels function as coincidence detectors for extracellular protons and other, as yet unidentified, endogenous factors. Purified MitTx elicits robust pain-related behaviour in mice by activation of ASIC1 channels on capsaicin-sensitive nerve fibres. These findings reveal a mechanism whereby snake venoms produce pain, and highlight an unexpected contribution of ASIC1 channels to nociception.


Asunto(s)
Venenos Elapídicos/química , Venenos Elapídicos/farmacología , Elapidae , Proteínas del Tejido Nervioso/metabolismo , Dolor/inducido químicamente , Multimerización de Proteína , Canales de Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Secuencia de Aminoácidos , Animales , Capsaicina/farmacología , Células Cultivadas , Miembro Posterior/efectos de los fármacos , Miembro Posterior/fisiopatología , Humanos , Concentración de Iones de Hidrógeno , Activación del Canal Iónico/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Oocitos , Dolor/metabolismo , Dolor/fisiopatología , Estructura Cuaternaria de Proteína , Protones , Ratas , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Agonistas de los Canales de Sodio , Canales de Sodio/deficiencia , Canales de Sodio/genética , Canales Catiónicos TRPV/metabolismo , Xenopus laevis
8.
Pain ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39073407

RESUMEN

ABSTRACT: Microglia take on an altered morphology during chronic opioid treatment. This morphological change is broadly used to identify the activated microglial state associated with opioid side effects, including tolerance and opioid-induced hyperalgesia (OIH). Microglia display similar morphological responses in the spinal cord after peripheral nerve injury (PNI). Consistent with this observation, functional studies have suggested that microglia activated by opioids or PNI engage common molecular mechanisms to induce hypersensitivity. In this article, we conducted deep RNA sequencing (RNA-seq) and morphological analysis of spinal cord microglia in male mice to comprehensively interrogate transcriptional states and mechanistic commonality between multiple models of OIH and PNI. After PNI, we identify an early proliferative transcriptional event across models that precedes the upregulation of histological markers of microglial activation. However, we found no proliferative transcriptional response associated with opioid-induced microglial activation, consistent with histological data, indicating that the number of microglia remains stable during morphine treatment, whereas their morphological response differs from PNI models. Collectively, these results establish the diversity of pain-associated microglial transcriptomic responses and point towards the targeting of distinct insult-specific microglial responses to treat OIH, PNI, or other central nervous system pathologies.

9.
Nat Neurosci ; 26(3): 430-446, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36732642

RESUMEN

Complex diseases are characterized by spatiotemporal cellular and molecular changes that may be difficult to comprehensively capture. However, understanding the spatiotemporal dynamics underlying pathology can shed light on disease mechanisms and progression. Here we introduce STARmap PLUS, a method that combines high-resolution spatial transcriptomics with protein detection in the same tissue section. As proof of principle, we analyze brain tissues of a mouse model of Alzheimer's disease at 8 and 13 months of age. Our approach provides a comprehensive cellular map of disease progression. It reveals a core-shell structure where disease-associated microglia (DAM) closely contact amyloid-ß plaques, whereas disease-associated astrocyte-like (DAA-like) cells and oligodendrocyte precursor cells (OPCs) are enriched in the outer shells surrounding the plaque-DAM complex. Hyperphosphorylated tau emerges mainly in excitatory neurons in the CA1 region and correlates with the local enrichment of oligodendrocyte subtypes. The STARmap PLUS method bridges single-cell gene expression profiles with tissue histopathology at subcellular resolution, providing a tool to pinpoint the molecular and cellular changes underlying pathology.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Péptidos beta-Amiloides , Astrocitos , Placa Amiloide , Precursor de Proteína beta-Amiloide , Ratones Transgénicos , Encéfalo
10.
Cell Rep ; 40(8): 111189, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-36001972

RESUMEN

Oligodendrocyte dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, so understanding oligodendrocyte activation states would shed light on disease processes. We identify three distinct activation states of oligodendrocytes from single-cell RNA sequencing (RNA-seq) of mouse models of Alzheimer's disease (AD) and multiple sclerosis (MS): DA1 (disease-associated1, associated with immunogenic genes), DA2 (disease-associated2, associated with genes influencing survival), and IFN (associated with interferon response genes). Spatial analysis of disease-associated oligodendrocytes (DAOs) in the cuprizone model reveals that DA1 and DA2 are established outside of the lesion area during demyelination and that DA1 repopulates the lesion during remyelination. Independent meta-analysis of human single-nucleus RNA-seq datasets reveals that the transcriptional responses of MS oligodendrocytes share features with mouse models. In contrast, the oligodendrocyte activation signature observed in human AD is largely distinct from those observed in mice. This catalog of oligodendrocyte activation states (http://research-pub.gene.com/OligoLandscape/) will be important to understand disease progression and develop therapeutic interventions.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Enfermedades Neurodegenerativas , Animales , Cuprizona/uso terapéutico , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Oligodendroglía
11.
Archaea ; 2011: 891531, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22162665

RESUMEN

RNase P is a highly conserved ribonucleoprotein enzyme that represents a model complex for understanding macromolecular RNA-protein interactions. Archaeal RNase P consists of one RNA and up to five proteins (Pop5, RPP30, RPP21, RPP29, and RPP38/L7Ae). Four of these proteins function in pairs (Pop5-RPP30 and RPP21-RPP29). We have used nuclear magnetic resonance (NMR) spectroscopy and isothermal titration calorimetry (ITC) to characterize the interaction between Pop5 and RPP30 from the hyperthermophilic archaeon Pyrococcus furiosus (Pfu). NMR backbone resonance assignments of free RPP30 (25 kDa) indicate that the protein is well structured in solution, with a secondary structure matching that observed in a closely related crystal structure. Chemical shift perturbations upon the addition of Pop5 (14 kDa) reveal its binding surface on RPP30. ITC experiments confirm a net 1 : 1 stoichiometry for this tight protein-protein interaction and exhibit complex isotherms, indicative of higher-order binding. Indeed, light scattering and size exclusion chromatography data reveal the complex to exist as a 78 kDa heterotetramer with two copies each of Pop5 and RPP30. These results will inform future efforts to elucidate the functional role of the Pop5-RPP30 complex in RNase P assembly and catalysis.


Asunto(s)
Multimerización de Proteína , Pyrococcus furiosus/enzimología , Ribonucleasa P/metabolismo , Calorimetría , Espectroscopía de Resonancia Magnética , Subunidades de Proteína/metabolismo
12.
Cell Rep ; 37(13): 110158, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965428

RESUMEN

Non-neuronal responses in neurodegenerative disease have received increasing attention as important contributors to disease pathogenesis and progression. Here we utilize single-cell RNA sequencing to broadly profile 13 cell types in three different mouse models of Alzheimer disease (AD), capturing the effects of tau-only, amyloid-only, or combined tau-amyloid pathology. We highlight microglia, oligodendrocyte, astrocyte, and T cell responses and compare them across these models. Notably, we identify two distinct transcriptional states for oligodendrocytes emerging differentially across disease models, and we determine their spatial distribution. Furthermore, we explore the impact of Trem2 deletion in the context of combined pathology. Trem2 knockout mice exhibit severely blunted microglial responses to combined tau and amyloid pathology, but responses from non-microglial cell types (oligodendrocytes, astrocytes, and T cells) are relatively unchanged. These results delineate core transcriptional states that are engaged in response to AD pathology, and how they are influenced by a key AD risk gene, Trem2.


Asunto(s)
Enfermedad de Alzheimer/patología , Amiloide/química , Astrocitos/patología , Glicoproteínas de Membrana/fisiología , Oligodendroglía/patología , Receptores Inmunológicos/fisiología , Linfocitos T/inmunología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodendroglía/inmunología , Oligodendroglía/metabolismo
13.
Neuron ; 109(8): 1283-1301.e6, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33675684

RESUMEN

Loss-of-function TREM2 mutations strongly increase Alzheimer's disease (AD) risk. Trem2 deletion has revealed protective Trem2 functions in preclinical models of ß-amyloidosis, a prominent feature of pre-diagnosis AD stages. How TREM2 influences later AD stages characterized by tau-mediated neurodegeneration is unclear. To understand Trem2 function in the context of both ß-amyloid and tau pathologies, we examined Trem2 deficiency in the pR5-183 mouse model expressing mutant tau alone or in TauPS2APP mice, in which ß-amyloid pathology exacerbates tau pathology and neurodegeneration. Single-cell RNA sequencing in these models revealed robust disease-associated microglia (DAM) activation in TauPS2APP mice that was amyloid-dependent and Trem2-dependent. In the presence of ß-amyloid pathology, Trem2 deletion further exacerbated tau accumulation and spreading and promoted brain atrophy. Without ß-amyloid pathology, Trem2 deletion did not affect these processes. Therefore, TREM2 may slow AD progression and reduce tau-driven neurodegeneration by restricting the degree to which ß-amyloid facilitates the spreading of pathogenic tau.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Atrofia/genética , Atrofia/metabolismo , Atrofia/patología , Encéfalo/patología , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Receptores Inmunológicos/genética , Proteínas tau/genética
14.
J Exp Med ; 217(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32357367

RESUMEN

Myeloid cells play critical and diverse roles in mammalian physiology, including tissue development and repair, innate defense against pathogens, and generation of adaptive immunity. As cells that show prolonged recruitment to sites of injury or pathology, myeloid cells represent therapeutic targets for a broad range of diseases. However, few approaches have been developed for gene editing of these cell types, likely owing to their sensitivity to foreign genetic material or virus-based manipulation. Here we describe optimized strategies for gene disruption in primary myeloid cells of human and murine origin. Using nucleofection-based delivery of Cas9-ribonuclear proteins (RNPs), we achieved near population-level genetic knockout of single and multiple targets in a range of cell types without selection or enrichment. Importantly, we show that cellular fitness and response to immunological stimuli is not significantly impacted by the gene editing process. This provides a significant advance in the study of myeloid cell biology, thus enabling pathway discovery and drug target validation across species in the field of innate immunity.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Inactivación de Genes , Técnicas de Transferencia de Gen , Células Mieloides/metabolismo , Animales , Células Cultivadas , Células Dendríticas/metabolismo , Eliminación de Gen , Edición Génica , Ingeniería Genética , Genoma , Humanos , Macrófagos/metabolismo , Ratones , Monocitos/metabolismo , Fagocitosis , Fenotipo , ARN Guía de Kinetoplastida/genética , Ribonucleoproteínas/metabolismo , Virus/metabolismo
15.
Curr Protoc Immunol ; 125(1): e70, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30414379

RESUMEN

Microglia represent 5-10% of cells in the central nervous system and contribute to the development, homeostasis, injury, and repair of neural tissues. As the tissue-resident macrophages of the central nervous system, microglia execute core innate immune functions such as detection of pathogens/damage, cytokine secretion, and phagocytosis. However, additional properties that are specific to microglia and their neural environment are beginning to be appreciated. This article describes approaches for purification of microglia by fluorescence-activated cell sorting using microglia-specific surface markers and for enrichment of microglia by magnetic sorting and immunopanning. Detailed information about culturing primary microglia at various developmental stages is also provided. Throughout, we focus on special considerations for handling microglia and compare the relative strengths or disadvantages of different protocols. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Técnicas de Cultivo de Célula , Citometría de Flujo , Microglía , Animales , Ratones , Ratas
16.
Cell Rep ; 27(10): 2895-2908.e4, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167136

RESUMEN

Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.


Asunto(s)
Encéfalo/diagnóstico por imagen , AMP Cíclico/metabolismo , Microglía/metabolismo , Seudópodos/metabolismo , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microtúbulos/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Seudópodos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal
17.
J Vis Exp ; (133)2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29578519

RESUMEN

Microglia represent 5 - 10% of all central nervous system (CNS) cells and are increasingly drawing attention due to their contributions during development, homeostasis, and disease. Although macrophages have been studied in detail for decades, specialized features of microglia, the tissue-resident macrophages of the CNS, have remained largely mysterious, in part due to limitations in the ability to recapitulate mature microglial properties in culture. Here, we illustrate a straightforward procedure for the rapid isolation of pure microglia from the mature rodent brain. We also describe serum-free culture conditions that support high levels of microglial viability over time. Microglia cultured under these defined-medium conditions exhibit elaborate ramified processes and dynamic surveillance behavior. We illustrate some effects of serum exposure on cultured microglia and discuss how these serum-free cultures compare to both serum-exposed cultures as well as microglia in vivo.


Asunto(s)
Medios de Cultivo Condicionados/metabolismo , Microglía/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Microglía/citología , Roedores
18.
Nat Genet ; 50(12): 1716-1727, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30397336

RESUMEN

Phagocytosis is required for a broad range of physiological functions, from pathogen defense to tissue homeostasis, but the mechanisms required for phagocytosis of diverse substrates remain incompletely understood. Here, we developed a rapid magnet-based phenotypic screening strategy, and performed eight genome-wide CRISPR screens in human cells to identify genes regulating phagocytosis of distinct substrates. After validating select hits in focused miniscreens, orthogonal assays and primary human macrophages, we show that (1) the previously uncharacterized gene NHLRC2 is a central player in phagocytosis, regulating RhoA-Rac1 signaling cascades that control actin polymerization and filopodia formation, (2) very-long-chain fatty acids are essential for efficient phagocytosis of certain substrates and (3) the previously uncharacterized Alzheimer's disease-associated gene TM2D3 can preferentially influence uptake of amyloid-ß aggregates. These findings illuminate new regulators and core principles of phagocytosis, and more generally establish an efficient method for unbiased identification of cellular uptake mechanisms across diverse physiological and pathological contexts.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Magnetismo/métodos , Fagocitosis/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulación de la Expresión Génica , Estudios de Asociación Genética/métodos , Genoma Humano , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Ratones , Células RAW 264.7 , Transducción de Señal/genética , Células U937
19.
Neuron ; 94(4): 759-773.e8, 2017 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-28521131

RESUMEN

Microglia, the resident macrophages of the CNS, engage in various CNS-specific functions that are critical for development and health. To better study microglia and the properties that distinguish them from other tissue macrophage populations, we have optimized serum-free culture conditions to permit robust survival of highly ramified adult microglia under defined-medium conditions. We find that astrocyte-derived factors prevent microglial death ex vivo and that this activity results from three primary components, CSF-1/IL-34, TGF-ß2, and cholesterol. Using microglial cultures that have never been exposed to serum, we demonstrate a dramatic and lasting change in phagocytic capacity after serum exposure. Finally, we find that mature microglia rapidly lose signature gene expression after isolation, and that this loss can be reversed by engrafting cells back into an intact CNS environment. These data indicate that the specialized gene expression profile of mature microglia requires continuous instructive signaling from the intact CNS.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Colesterol/farmacología , Interleucinas/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , Animales , Astrocitos/metabolismo , Técnicas de Cultivo de Célula , Colesterol/metabolismo , Medios de Cultivo Condicionados/metabolismo , Humanos , Interleucinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones , Microglía/citología , Microglía/inmunología , Microglía/metabolismo , Fagocitosis/inmunología , Ratas , Suero , Transcriptoma , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta2/metabolismo
20.
Nat Med ; 23(2): 164-173, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28092666

RESUMEN

Opioid pain medications have detrimental side effects including analgesic tolerance and opioid-induced hyperalgesia (OIH). Tolerance and OIH counteract opioid analgesia and drive dose escalation. The cell types and receptors on which opioids act to initiate these maladaptive processes remain disputed, which has prevented the development of therapies to maximize and sustain opioid analgesic efficacy. We found that µ opioid receptors (MORs) expressed by primary afferent nociceptors initiate tolerance and OIH development. RNA sequencing and histological analysis revealed that MORs are expressed by nociceptors, but not by spinal microglia. Deletion of MORs specifically in nociceptors eliminated morphine tolerance, OIH and pronociceptive synaptic long-term potentiation without altering antinociception. Furthermore, we found that co-administration of methylnaltrexone bromide, a peripherally restricted MOR antagonist, was sufficient to abrogate morphine tolerance and OIH without diminishing antinociception in perioperative and chronic pain models. Collectively, our data support the idea that opioid agonists can be combined with peripheral MOR antagonists to limit analgesic tolerance and OIH.


Asunto(s)
Analgésicos Opioides/farmacología , Tolerancia a Medicamentos/genética , Hiperalgesia/genética , Microglía/metabolismo , Morfina/farmacología , Nociceptores/metabolismo , Receptores Opioides mu/genética , Analgesia , Animales , Dolor Crónico , Modelos Animales de Enfermedad , Eliminación de Gen , Hiperalgesia/inducido químicamente , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Ratones , Ratones Noqueados , Naltrexona/análogos & derivados , Naltrexona/farmacología , Nocicepción/efectos de los fármacos , Dolor Postoperatorio , Compuestos de Amonio Cuaternario/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Transducción de Señal , Médula Espinal/citología , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA