Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 180(3): 502-520.e19, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31983537

RESUMEN

The tumor microenvironment (TME) is critical for tumor progression. However, the establishment and function of the TME remain obscure because of its complex cellular composition. Using a mouse genetic system called mosaic analysis with double markers (MADMs), we delineated TME evolution at single-cell resolution in sonic hedgehog (SHH)-activated medulloblastomas that originate from unipotent granule neuron progenitors in the brain. First, we found that astrocytes within the TME (TuAstrocytes) were trans-differentiated from tumor granule neuron precursors (GNPs), which normally never differentiate into astrocytes. Second, we identified that TME-derived IGF1 promotes tumor progression. Third, we uncovered that insulin-like growth factor 1 (IGF1) is produced by tumor-associated microglia in response to interleukin-4 (IL-4) stimulation. Finally, we found that IL-4 is secreted by TuAstrocytes. Collectively, our studies reveal an evolutionary process that produces a multi-lateral network within the TME of medulloblastoma: a fraction of tumor cells trans-differentiate into TuAstrocytes, which, in turn, produce IL-4 that stimulates microglia to produce IGF1 to promote tumor progression.


Asunto(s)
Astrocitos/metabolismo , Carcinogénesis/metabolismo , Transdiferenciación Celular , Neoplasias Cerebelosas/metabolismo , Meduloblastoma/metabolismo , Comunicación Paracrina , Animales , Linaje de la Célula , Neoplasias Cerebelosas/patología , Modelos Animales de Enfermedad , Femenino , Proteínas Hedgehog/metabolismo , Xenoinjertos , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Microambiente Tumoral
3.
Immunity ; 48(5): 1014-1028.e6, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752062

RESUMEN

Stromal cells (SCs) establish the compartmentalization of lymphoid tissues critical to the immune response. However, the full diversity of lymph node (LN) SCs remains undefined. Using droplet-based single-cell RNA sequencing, we identified nine peripheral LN non-endothelial SC clusters. Included are the established subsets, Ccl19hi T-zone reticular cells (TRCs), marginal reticular cells, follicular dendritic cells (FDCs), and perivascular cells. We also identified Ccl19lo TRCs, likely including cholesterol-25-hydroxylase+ cells located at the T-zone perimeter, Cxcl9+ TRCs in the T-zone and interfollicular region, CD34+ SCs in the capsule and medullary vessel adventitia, indolethylamine N-methyltransferase+ SCs in the medullary cords, and Nr4a1+ SCs in several niches. These data help define how transcriptionally distinct LN SCs support niche-restricted immune functions and provide evidence that many SCs are in an activated state.


Asunto(s)
Ganglios Linfáticos/inmunología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Células del Estroma/inmunología , Transcriptoma/inmunología , Animales , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL19/metabolismo , Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/metabolismo , Femenino , Ganglios Linfáticos/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones Endogámicos C57BL , Células del Estroma/metabolismo
4.
Semin Immunol ; 60: 101650, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-36099864

RESUMEN

Viral infections of the central nervous system (CNS) are a significant cause of neurological impairment and mortality worldwide. As tissue resident macrophages, microglia are critical initial responders to CNS viral infection. Microglia seem to coordinate brain-wide antiviral responses of both brain resident cells and infiltrating immune cells. This review discusses how microglia may promote this antiviral response at a molecular level, from potential mechanisms of virus recognition to downstream cytokine responses and interaction with antiviral T cells. Recent advancements in genetic tools to specifically target microglia in vivo promise to further our understanding about the precise mechanistic role of microglia in CNS infection.


Asunto(s)
Antivirales , Microglía , Humanos , Encéfalo , Médula Espinal , Sistema Nervioso Central
5.
EMBO J ; 39(16): e105924, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32705698

RESUMEN

Microglia, the brain's tissue-resident macrophages, contribute to the developmental elimination of extranumerary synapses and to pathologic synapse loss in mouse models of neurodegeneration. Two papers published in The EMBO Journal reveal that phosphatidylserine (PS) is a neuronal cue for microglial synapse elimination.


Asunto(s)
Microglía , Fosfatidilserinas , Animales , Apoptosis , Señales (Psicología) , Ratones , Isoformas de Proteínas , Receptores Acoplados a Proteínas G , Sinapsis
6.
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
7.
Proc Natl Acad Sci U S A ; 116(3): 997-1006, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30602457

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive malignant brain tumor with fatal outcome. Tumor-associated macrophages and microglia (TAMs) have been found to be major tumor-promoting immune cells in the tumor microenvironment. Hence, modulation and reeducation of tumor-associated macrophages and microglia in GBM is considered a promising antitumor strategy. Resident microglia and invading macrophages have been shown to have distinct origin and function. Whereas yolk sac-derived microglia reside in the brain, blood-derived monocytes invade the central nervous system only under pathological conditions like tumor formation. We recently showed that disruption of the SIRPα-CD47 signaling axis is efficacious against various brain tumors including GBM primarily by inducing tumor phagocytosis. However, most effects are attributed to macrophages recruited from the periphery but the role of the brain resident microglia is unknown. Here, we sought to utilize a model to distinguish resident microglia and peripheral macrophages within the GBM-TAM pool, using orthotopically xenografted, immunodeficient, and syngeneic mouse models with genetically color-coded macrophages (Ccr2RFP) and microglia (Cx3cr1GFP). We show that even in the absence of phagocytizing macrophages (Ccr2RFP/RFP), microglia are effector cells of tumor cell phagocytosis in response to anti-CD47 blockade. Additionally, macrophages and microglia show distinct morphological and transcriptional changes. Importantly, the transcriptional profile of microglia shows less of an inflammatory response which makes them a promising target for clinical applications.


Asunto(s)
Neoplasias Encefálicas/inmunología , Antígeno CD47/inmunología , Glioblastoma/inmunología , Microglía/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/inmunología , Fagocitosis , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Animales , Neoplasias Encefálicas/patología , Antígeno CD47/genética , Glioblastoma/genética , Glioblastoma/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Microglía/patología , Monocitos/inmunología , Monocitos/patología , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptores Inmunológicos/genética , Transducción de Señal/genética
8.
Neurobiol Dis ; 148: 105172, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33171230

RESUMEN

In injury and disease, microglia and astrocytes - two major non-neuronal cell types in the central nervous system (CNS) - undergo morphological, transcriptional, and functional changes, which can underlie pathogenesis and dysfunction of the CNS. Microglia, the brain's tissue resident parenchymal macrophages, are described as becoming "activated" as they deftly change their production of different inflammatory mediators, alter the surveillance behavior of their cellular protrusions, and differentially influence the function of astrocytes. For their part, astrocytes - the most abundant glial cell type - are said to become "reactive", which implies (perhaps inappropriately) causality for the changes astrocytes undergo. Reactive astrocytes variably undergo process hypertrophy, decrease their normal homeostatic functions such as facilitating synapse formation, and in some cases act to form a tissue scar in response to insult. But what do these terms "activation" and "reactivity" mean, anyway? And how do these changed microglia and astrocytes contribute to neurodegenerative disease (ND)? Here, we describe our current understanding of the role of activated and reactive microglia and astrocytes in ND, as well as our current understanding about what these states are and might mean. We survey the earliest description of these cells by histopathologists, their transcriptomic identities, and finally our mechanistic understanding of their functions in ND.


Asunto(s)
Astrocitos/metabolismo , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Astrocitos/patología , Humanos , Microglía/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/metabolismo , Neuroglía/patología
9.
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
10.
Nature ; 486(7403): 410-4, 2012 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-22722203

RESUMEN

In the developing central nervous system (CNS), the control of synapse number and function is critical to the formation of neural circuits. We previously demonstrated that astrocyte-secreted factors powerfully induce the formation of functional excitatory synapses between CNS neurons. Astrocyte-secreted thrombospondins induce the formation of structural synapses, but these synapses are postsynaptically silent. Here we use biochemical fractionation of astrocyte-conditioned medium to identify glypican 4 (Gpc4) and glypican 6 (Gpc6) as astrocyte-secreted signals sufficient to induce functional synapses between purified retinal ganglion cell neurons, and show that depletion of these molecules from astrocyte-conditioned medium significantly reduces its ability to induce postsynaptic activity. Application of Gpc4 to purified neurons is sufficient to increase the frequency and amplitude of glutamatergic synaptic events. This is achieved by increasing the surface level and clustering, but not overall cellular protein level, of the GluA1 subunit of the AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) glutamate receptor (AMPAR). Gpc4 and Gpc6 are expressed by astrocytes in vivo in the developing CNS, with Gpc4 expression enriched in the hippocampus and Gpc6 enriched in the cerebellum. Finally, we demonstrate that Gpc4-deficient mice have defective synapse formation, with decreased amplitude of excitatory synaptic currents in the developing hippocampus and reduced recruitment of AMPARs to synapses. These data identify glypicans as a family of novel astrocyte-derived molecules that are necessary and sufficient to promote glutamate receptor clustering and receptivity and to induce the formation of postsynaptically functioning CNS synapses.


Asunto(s)
Astrocitos/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Glipicanos/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Animales , Astrocitos/citología , Cerebelo/citología , Cerebelo/metabolismo , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Femenino , Glipicanos/deficiencia , Glipicanos/farmacología , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/patología
11.
EMBO J ; 36(22): 3269-3271, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29101295
12.
J Neurosci ; 34(36): 11929-47, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25186741

RESUMEN

The major cell classes of the brain differ in their developmental processes, metabolism, signaling, and function. To better understand the functions and interactions of the cell types that comprise these classes, we acutely purified representative populations of neurons, astrocytes, oligodendrocyte precursor cells, newly formed oligodendrocytes, myelinating oligodendrocytes, microglia, endothelial cells, and pericytes from mouse cerebral cortex. We generated a transcriptome database for these eight cell types by RNA sequencing and used a sensitive algorithm to detect alternative splicing events in each cell type. Bioinformatic analyses identified thousands of new cell type-enriched genes and splicing isoforms that will provide novel markers for cell identification, tools for genetic manipulation, and insights into the biology of the brain. For example, our data provide clues as to how neurons and astrocytes differ in their ability to dynamically regulate glycolytic flux and lactate generation attributable to unique splicing of PKM2, the gene encoding the glycolytic enzyme pyruvate kinase. This dataset will provide a powerful new resource for understanding the development and function of the brain. To ensure the widespread distribution of these datasets, we have created a user-friendly website (http://web.stanford.edu/group/barres_lab/brain_rnaseq.html) that provides a platform for analyzing and comparing transciption and alternative splicing profiles for various cell classes in the brain.


Asunto(s)
Empalme Alternativo , Corteza Cerebral/metabolismo , Bases de Datos de Ácidos Nucleicos , Endotelio Vascular/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Transcriptoma , Animales , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/citología , Ratones , Análisis de Secuencia de ARN
13.
AJNR Am J Neuroradiol ; 45(6): 769-772, 2024 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-38697787

RESUMEN

BACKGROUND AND PURPOSE: While classic brain MR imaging features of Alexander disease have been well-documented, lesional patterns can overlap with other leukodystrophies, especially in the early stages of the disease or in milder phenotypes. We aimed to assess the utility of a new neuroimaging sign to help increase the diagnostic specificity of Alexander disease. MATERIALS AND METHODS: A peculiar bilateral symmetric hyperintense signal on T2-weighted images affecting the medulla oblongata was identified in an index patient with type I Alexander disease. Subsequently, 5 observers performed a systematic MR imaging review for this pattern by examining 55 subjects with Alexander disease and 74 subjects with other leukodystrophies. Interobserver agreement was assessed by the κ index. Sensitivity, specificity, and receiver operating characteristic curves were determined. RESULTS: The identified pattern was present in 87% of subjects with Alexander disease and 14% of those without Alexander disease leukodystrophy (P < .001), 3 with vanishing white matter, 4 with adult polyglucosan body disease, and 3 others. It was found equally in both type I and type II Alexander disease (28/32, 88% versus 18/21, 86%; P = .851) and in subjects with unusual disease features (2/2). Sensitivity (87.3%; 95% CI, 76.0%-93.7%), specificity (86.5%; 95% CI, 76.9%-92.5%), and interobserver agreement (κ index = 0.82) were high. CONCLUSIONS: The identified pattern in the medulla oblongata, called the chipmunk sign due to its resemblance to the face of this rodent, is extremely common in subjects with Alexander disease and represents a diagnostic tool that can aid in early diagnosis, especially in subjects with otherwise atypical MR imaging findings and/or clinical features.


Asunto(s)
Enfermedad de Alexander , Imagen por Resonancia Magnética , Sensibilidad y Especificidad , Humanos , Enfermedad de Alexander/diagnóstico por imagen , Masculino , Femenino , Adulto , Imagen por Resonancia Magnética/métodos , Persona de Mediana Edad , Adulto Joven , Adolescente , Tronco Encefálico/diagnóstico por imagen , Tronco Encefálico/patología , Niño , Anciano , Bulbo Raquídeo/diagnóstico por imagen , Bulbo Raquídeo/patología , Preescolar
14.
J Clin Neurophysiol ; 40(2): e6-e9, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36308754

RESUMEN

SUMMARY: Quantitative analysis of continuous electroencephalography (QEEG) is increasingly being used to augment seizure detection in critically ill patients. Typically, seizures manifest on QEEG as abrupt increases in power and frequency, a visual pattern often called "flames." Here, we present a case of a 16-year-old patient with intractable Lennox-Gastaut syndrome secondary to a pathogenic variant in the SCN2A gene who had tonic seizures that manifest as abrupt decreases in power on QEEG, a visual pattern we term "icicles." Recognition of QEEG patterns representative of different seizure types is important as QEEG use becomes more widespread including in pediatric populations.


Asunto(s)
Síndrome de Lennox-Gastaut , Niño , Humanos , Adolescente , Síndrome de Lennox-Gastaut/diagnóstico , Síndrome de Lennox-Gastaut/complicaciones , Convulsiones/diagnóstico , Convulsiones/complicaciones , Electroencefalografía
15.
J Exp Med ; 220(3)2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36584406

RESUMEN

Hematopoietic stem cell transplantation (HSCT) can replace endogenous microglia with circulation-derived macrophages but has high mortality. To mitigate the risks of HSCT and expand the potential for microglia replacement, we engineered an inhibitor-resistant CSF1R that enables robust microglia replacement. A glycine to alanine substitution at position 795 of human CSF1R (G795A) confers resistance to multiple CSF1R inhibitors, including PLX3397 and PLX5622. Biochemical and cell-based assays show no discernable gain or loss of function. G795A- but not wildtype-CSF1R expressing macrophages efficiently engraft the brain of PLX3397-treated mice and persist after cessation of inhibitor treatment. To gauge translational potential, we CRISPR engineered human-induced pluripotent stem cell-derived microglia (iMG) to express G795A. Xenotransplantation studies demonstrate that G795A-iMG exhibit nearly identical gene expression to wildtype iMG, respond to inflammatory stimuli, and progressively expand in the presence of PLX3397, replacing endogenous microglia to fully occupy the brain. In sum, we engineered a human CSF1R variant that enables nontoxic, cell type, and tissue-specific replacement of microglia.


Asunto(s)
Microglía , Ingeniería de Proteínas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Humanos , Ratones , Aminopiridinas/farmacología , Encéfalo/metabolismo , Microglía/metabolismo , Ingeniería de Proteínas/métodos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos
16.
Nat Commun ; 14(1): 5632, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37704594

RESUMEN

With concurrent global epidemics of chronic pain and opioid use disorders, there is a critical need to identify, target and manipulate specific cell populations expressing the mu-opioid receptor (MOR). However, available tools and transgenic models for gaining long-term genetic access to MOR+ neural cell types and circuits involved in modulating pain, analgesia and addiction across species are limited. To address this, we developed a catalog of MOR promoter (MORp) based constructs packaged into adeno-associated viral vectors that drive transgene expression in MOR+ cells. MORp constructs designed from promoter regions upstream of the mouse Oprm1 gene (mMORp) were validated for transduction efficiency and selectivity in endogenous MOR+ neurons in the brain, spinal cord, and periphery of mice, with additional studies revealing robust expression in rats, shrews, and human induced pluripotent stem cell (iPSC)-derived nociceptors. The use of mMORp for in vivo fiber photometry, behavioral chemogenetics, and intersectional genetic strategies is also demonstrated. Lastly, a human designed MORp (hMORp) efficiently transduced macaque cortical OPRM1+ cells. Together, our MORp toolkit provides researchers cell type specific genetic access to target and functionally manipulate mu-opioidergic neurons across a range of vertebrate species and translational models for pain, addiction, and neuropsychiatric disorders.


Asunto(s)
Analgesia , Dolor Crónico , Células Madre Pluripotentes Inducidas , Animales , Humanos , Ratones , Ratas , Macaca , Receptores Opioides , Receptores Opioides mu/genética , Transgenes
17.
Cell Stem Cell ; 29(11): 1594-1610.e8, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36332572

RESUMEN

The molecular diversity of glia in the human hippocampus and their temporal dynamics over the lifespan remain largely unknown. Here, we performed single-nucleus RNA sequencing to generate a transcriptome atlas of the human hippocampus across the postnatal lifespan. Detailed analyses of astrocytes, oligodendrocyte lineages, and microglia identified subpopulations with distinct molecular signatures and revealed their association with specific physiological functions, age-dependent changes in abundance, and disease relevance. We further characterized spatiotemporal heterogeneity of GFAP-enriched astrocyte subpopulations in the hippocampal formation using immunohistology. Leveraging glial subpopulation classifications as a reference map, we revealed the diversity of glia differentiated from human pluripotent stem cells and identified dysregulated genes and pathological processes in specific glial subpopulations in Alzheimer's disease (AD). Together, our study significantly extends our understanding of human glial diversity, population dynamics across the postnatal lifespan, and dysregulation in AD and provides a reference atlas for stem-cell-based glial differentiation.


Asunto(s)
Enfermedad de Alzheimer , Transcriptoma , Humanos , Transcriptoma/genética , Longevidad/genética , Neuroglía/patología , Hipocampo , Astrocitos/patología , Enfermedad de Alzheimer/patología
18.
Cell Stem Cell ; 28(12): 2035-2036, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34861141

RESUMEN

Dissecting contributions of microglia to human brain development and disease pathogenesis requires modeling interactions between these microglia and their local environment. In this issue of Cell Stem Cell, Popova et al. (2021) propose a transcriptomic "microglia report card" and create a neuroimmune organoid to model complex interactions involving human microglia.


Asunto(s)
Microglía , Organoides , Encéfalo , Humanos , Transcriptoma/genética
19.
Nat Neurosci ; 23(2): 157-166, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792468

RESUMEN

Microglia are the tissue-resident macrophages of the brain and spinal cord. They are critical players in the development, normal function, and decline of the CNS. Unlike traditional monocyte-derived macrophages, microglia originate from primitive hematopoiesis in the embryonic yolk sac and self-renew throughout life. Microglia also have a unique genetic signature among tissue resident macrophages. Recent studies identify the contributions of both brain environment and developmental history to the transcriptomic identity of microglia. Here we review this emerging literature and discuss the potential implications of origin on microglial function, with particular focus on existing and future therapies using bone-marrow- or stem-cell-derived cells for the treatment of neurological diseases.


Asunto(s)
Encéfalo/citología , Microglía/citología , Animales , Linaje de la Célula/fisiología , Humanos , Macrófagos/citología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA