Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37882444

RESUMEN

During human forebrain development, neural progenitor cells (NPCs) in the ventricular zone (VZ) undergo asymmetric cell divisions to produce a self-renewed progenitor cell, maintaining the potential to go through additional rounds of cell divisions, and differentiating daughter cells, populating the developing cortex. Previous work in the embryonic rodent brain suggested that the preferential inheritance of the pre-existing (older) centrosome to the self-renewed progenitor cell is required to maintain stem cell properties, ensuring proper neurogenesis. If asymmetric segregation of centrosomes occurs in NPCs of the developing human brain, which depends on unique molecular regulators and species-specific cellular composition, remains unknown. Using a novel, recombination-induced tag exchange-based genetic tool to birthdate and track the segregation of centrosomes over multiple cell divisions in human embryonic stem cell-derived regionalised forebrain organoids, we show the preferential inheritance of the older mother centrosome towards self-renewed NPCs. Aberration of asymmetric segregation of centrosomes by genetic manipulation of the centrosomal, microtubule-associated protein Ninein alters fate decisions of NPCs and their maintenance in the VZ of human cortical organoids. Thus, the data described here use a novel genetic approach to birthdate centrosomes in human cells and identify asymmetric inheritance of centrosomes as a mechanism to maintain self-renewal properties and to ensure proper neurogenesis in human NPCs.


Asunto(s)
Células-Madre Neurales , Humanos , Centrosoma/metabolismo , División Celular , Encéfalo/metabolismo , Neurogénesis
2.
Am J Pathol ; 193(12): 2144-2155, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37741454

RESUMEN

Mechanisms responsible for the pathogenesis of diabetic retinal disease remain incompletely understood, but they likely involve multiple cellular targets, including photoreceptors. Evidence suggests that dysregulated de novo lipogenesis in photoreceptors is a critical early target of diabetes. Following on this observation, the present study aimed to determine whether two interventions shown to improve diabetic retinopathy in mice-pharmacologic visual cycle inhibition and prolonged dark adaptation-reduce photoreceptor anabolic lipid metabolism. Elevated retinal lipid biosynthetic signaling was observed in two mouse models of diabetes, with both models showing reduced retinal AMP-activated kinase (AMPK) signaling, elevated acetyl CoA carboxylase (ACC) signaling, and increased activity of fatty acid synthase, which promotes lipotoxicity in photoreceptors. Although retinal AMPK-ACC axis signaling was dependent on systemic glucose fluctuations in healthy animals, mice with diabetes lacked such regulation. Visual cycle inhibition and prolonged dark adaptation reversed abnormal retinal AMPK-ACC signaling in mice with diabetes. Although visual cycle inhibition reduced the severity of diabetic retinopathy in control mice, as assessed by retinal capillary atrophy, this intervention was ineffective in fatty acid synthase gain-of-function mice. These results suggest that early diabetic retinopathy is characterized by glucose-driven elevations in retinal lipid biosynthetic activity, and that two interventions known to increase photoreceptor glucose demands alleviate disease by reversing these signals.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Degeneración Retiniana , Ratones , Animales , Proteínas Quinasas Activadas por AMP/metabolismo , Retinopatía Diabética/metabolismo , Glucosa , Ácido Graso Sintasas , Lípidos
3.
EMBO J ; 42(17): e112847, 2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37365982

RESUMEN

The paralogs CUL4A and CUL4B assemble cullin-RING E3 ubiquitin ligase (CRL) complexes regulating multiple chromatin-associated cellular functions. Although they are structurally similar, we found that the unique N-terminal extension of CUL4B is heavily phosphorylated during mitosis, and the phosphorylation pattern is perturbed in the CUL4B-P50L mutation causing X-linked intellectual disability (XLID). Phenotypic characterization and mutational analysis revealed that CUL4B phosphorylation is required for efficient progression through mitosis, controlling spindle positioning and cortical tension. While CUL4B phosphorylation triggers chromatin exclusion, it promotes binding to actin regulators and to two previously unrecognized CUL4B-specific substrate receptors (DCAFs), LIS1 and WDR1. Indeed, co-immunoprecipitation experiments and biochemical analysis revealed that LIS1 and WDR1 interact with DDB1, and their binding is enhanced by the phosphorylated N-terminal domain of CUL4B. Finally, a human forebrain organoid model demonstrated that CUL4B is required to develop stable ventricular structures that correlate with onset of forebrain differentiation. Together, our study uncovers previously unrecognized DCAFs relevant for mitosis and brain development that specifically bind CUL4B, but not the CUL4B-P50L patient mutant, by a phosphorylation-dependent mechanism.


Asunto(s)
Mitosis , Ubiquitina-Proteína Ligasas , Humanos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Cromatina , Encéfalo/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo
4.
Nat Aging ; 3(4): 380-390, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37117787

RESUMEN

Neural stem cells (NSCs) generate new neurons throughout life in the mammalian hippocampus1. Advancing age leads to a decline in neurogenesis, which is associated with impaired cognition2,3. The cellular mechanisms causing reduced neurogenesis with advancing age remain largely unknown. We genetically labeled NSCs through conditional recombination driven by the regulatory elements of the stem-cell-expressed gene GLI family zinc finger 1 (Gli1) and used chronic intravital imaging to follow individual NSCs and their daughter cells over months within their hippocampal niche4,5. We show that aging affects multiple steps, from cell cycle entry of quiescent NSCs to determination of the number of surviving cells, ultimately causing reduced clonal output of individual NSCs. Thus, we here define the developmental stages that may be targeted to enhance neurogenesis with the aim of maintaining hippocampal plasticity with advancing age.


Asunto(s)
Disfunción Cognitiva , Células-Madre Neurales , Ratones , Animales , Neuronas/metabolismo , Neurogénesis/fisiología , Hipocampo , Disfunción Cognitiva/metabolismo , Mamíferos
5.
Hippocampus ; 33(4): 402-411, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36256493

RESUMEN

Neural stem cells (NSCs) generate new neurons throughout life in the mammalian hippocampus. The distinct developmental steps in the course of adult neurogenesis, including NSC activation, expansion, and neuronal integration, are increasingly well characterized down to the molecular level. However, substantial gaps remain in our knowledge about regulators and mechanisms involved in this biological process. This review highlights three long-standing unknowns. First, we discuss potency and identity of NSCs and the quest for a unifying model of short- and long-term self-renewal dynamics. Next, we examine cell death, specifically focusing on the early demise of newborn cells. Then, we outline the current knowledge on cell integration dynamics, discussing which (if any) neurons are replaced by newly added neurons in the hippocampal circuits. For each of these unknowns, we summarize the trajectory of studies leading to the current state of knowledge. Finally, we offer suggestions on how to fill the remaining gaps by taking advantage of novel technology to reveal currently hidden secrets in the course of adult hippocampal neurogenesis.


Asunto(s)
Hipocampo , Células-Madre Neurales , Adulto , Humanos , Animales , Recién Nacido , Hipocampo/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Células-Madre Neurales/fisiología , Mamíferos
6.
Development ; 149(20)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35815653

RESUMEN

Asymmetric segregation of cellular components regulates the fate and behavior of somatic stem cells. Similar to dividing budding yeast and precursor cells in Caenorhabditis elegans, it has been shown that mouse neural progenitors establish a diffusion barrier in the membrane of the endoplasmic reticulum (ER), which has been associated with asymmetric partitioning of damaged proteins and cellular age. However, the existence of an ER diffusion barrier in human cells remains unknown. Here, we used fluorescence loss in photobleaching (FLIP) imaging to show that human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived neural progenitor cells establish an ER diffusion barrier during cell division. The human ER diffusion barrier is regulated via lamin-dependent mechanisms and is associated with asymmetric segregation of mono- and polyubiquitylated damaged proteins. Further, forebrain regionalized organoids derived from hESCs were used to show the establishment of an ER membrane diffusion barrier in more naturalistic tissues, mimicking early steps of human brain development. Thus, the data provided here show that human neural progenitors establish a diffusion barrier during cell division in the membrane of the ER, which may allow for asymmetric segregation of cellular components, contributing to the fate and behavior of human neural progenitor cells.


Asunto(s)
División Celular , Retículo Endoplásmico , Células-Madre Neurales , Difusión , Retículo Endoplásmico/metabolismo , Humanos , Células-Madre Neurales/metabolismo
7.
STAR Protoc ; 3(1): 101129, 2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35118433

RESUMEN

Pluripotent stem cell-derived human cortical organoids allow for the analysis of stem cell behavior and neurogenesis in neural tissues. Delivery of plasmid DNA into organoids permits visualization of individual cells, characterization of cellular components, and manipulation of gene expression. We describe a protocol to transfect cells inside organoids with plasmid DNA using micro-injection and electroporation, allowing for DNA delivery to cells within cortical units. This protocol was optimized for cortical organoids; however, it may be adapted to other organoid models. For complete details on the use and execution of this protocol, please refer to Denoth-Lippuner et al. (2021).


Asunto(s)
Corteza Cerebral/metabolismo , ADN/genética , Electroporación , Organoides/metabolismo , Plásmidos , Humanos , Transfección
8.
Elife ; 112022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35089129

RESUMEN

Advancing age causes reduced hippocampal neurogenesis, associated with age-related cognitive decline. The spatial relationship of age-induced alterations in neural stem cells (NSCs) and surrounding cells within the hippocampal niche remains poorly understood due to limitations of antibody-based cellular phenotyping. We established iterative indirect immunofluorescence imaging (4i) in tissue sections, allowing for simultaneous detection of 18 proteins to characterize NSCs and surrounding cells in 2-, 6-, and 12-month-old mice. We show that reorganization of the dentate gyrus (DG) niche already occurs in middle-aged mice, paralleling the decline in neurogenesis. 4i-based tissue analysis of the DG identifies changes in cell-type contributions to the blood-brain barrier and microenvironments surrounding NSCs to play a pivotal role to preserve neurogenic permissiveness. The data provided represent a resource to characterize the principles causing alterations of stem cell-associated plasticity within the aging DG and provide a blueprint to analyze somatic stem cell niches across lifespan in complex tissues.


Asunto(s)
Envejecimiento , Giro Dentado/citología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Barrera Hematoencefálica , Encéfalo/embriología , Giro Dentado/diagnóstico por imagen , Giro Dentado/embriología , Giro Dentado/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Células Madre Embrionarias Humanas , Humanos , Masculino , Ratones Endogámicos C57BL , Organoides , Proteínas/análisis , Nicho de Células Madre
9.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34996870

RESUMEN

Fate and behavior of neural progenitor cells are tightly regulated during mammalian brain development. Metabolic pathways, such as glycolysis and oxidative phosphorylation, that are required for supplying energy and providing molecular building blocks to generate cells govern progenitor function. However, the role of de novo lipogenesis, which is the conversion of glucose into fatty acids through the multienzyme protein fatty acid synthase (FASN), for brain development remains unknown. Using Emx1Cre-mediated, tissue-specific deletion of Fasn in the mouse embryonic telencephalon, we show that loss of FASN causes severe microcephaly, largely due to altered polarity of apical, radial glia progenitors and reduced progenitor proliferation. Furthermore, genetic deletion and pharmacological inhibition of FASN in human embryonic stem cell-derived forebrain organoids identifies a conserved role of FASN-dependent lipogenesis for radial glia cell polarity in human brain organoids. Thus, our data establish a role of de novo lipogenesis for mouse and human brain development and identify a link between progenitor-cell polarity and lipid metabolism.


Asunto(s)
Encéfalo/metabolismo , Ácido Graso Sintasas/metabolismo , Lipogénesis/fisiología , Animales , Tipificación del Cuerpo , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Acido Graso Sintasa Tipo I , Ácido Graso Sintasas/genética , Humanos , Metabolismo de los Lípidos , Lipogénesis/genética , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Transcriptoma
10.
Cell Stem Cell ; 28(11): 2020-2034.e12, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34525348

RESUMEN

The division potential of individual stem cells and the molecular consequences of successive rounds of proliferation remain largely unknown. Here, we developed an inducible cell division counter (iCOUNT) that reports cell division events in human and mouse tissues in vitro and in vivo. Analyzing cell division histories of neural stem/progenitor cells (NSPCs) in the developing and adult brain, we show that iCOUNT can provide novel insights into stem cell behavior. Further, we use single-cell RNA sequencing (scRNA-seq) of iCOUNT-labeled NSPCs and their progenies from the developing mouse cortex and forebrain-regionalized human organoids to identify functionally relevant molecular pathways that are commonly regulated between mouse and human cells, depending on individual cell division histories. Thus, we developed a tool to characterize the molecular consequences of repeated cell divisions of stem cells that allows an analysis of the cellular principles underlying tissue formation, homeostasis, and repair.


Asunto(s)
Células-Madre Neurales , Animales , Encéfalo , División Celular , Proliferación Celular , Ratones , Organoides , Análisis de Secuencia de ARN
11.
STAR Protoc ; 2(3): 100695, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34382020

RESUMEN

This protocol describes the isolation and culturing of primary neural stem cells (NSCs) from the adult mouse hippocampus, followed by the experimental approach for fluorescence loss in photobleaching assays, previously used to characterize the presence of an endoplasmic reticulum (ER) membrane diffusion barrier. The assay described here can be used to study live asymmetry in the ER membrane or other organelles that is established in dividing NSCs. For complete details on the use and execution of this protocol, please refer to Clay et al. (2014); bin Imtiaz et al. (2021); Lee et al. (2016); Luedeke et al. (2005); Moore et al. (2015); Shcheprova et al. (2008).


Asunto(s)
Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Células-Madre Neurales/fisiología , Células Madre Adultas , Animales , Difusión , Retículo Endoplásmico/metabolismo , Fluorescencia , Proteínas Fluorescentes Verdes , Hipocampo/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Fotoblanqueo , Cultivo Primario de Células
12.
J Biol Chem ; 297(3): 101104, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34425110

RESUMEN

Diabetic retinopathy (DR) is an increasingly frequent cause of blindness across populations; however, the events that initiate pathophysiology of DR remain elusive. Strong preclinical and clinical evidence suggests that abnormalities in retinal lipid metabolism caused by diabetes may account for the origin of this disease. A major arm of lipid metabolism, de novo biosynthesis, is driven by elevation in available glucose, a common thread binding all forms of vision loss in diabetes. Therefore, we hypothesized that aberrant retinal lipid biogenesis is an important promoter of early DR. In murine models, we observed elevations of diabetes-associated retinal de novo lipogenesis ∼70% over control levels. This shift was primarily because of activation of fatty acid synthase (FAS), a rate-limiting enzyme in the biogenic pathway. Activation of FAS was driven by canonical glucose-mediated disinhibition of acetyl-CoA carboxylase, a major upstream regulatory enzyme. Mutant mice expressing gain-of-function FAS demonstrated increased vulnerability to DR, whereas those with FAS deletion in rod photoreceptors maintained preserved visual responses upon induction of diabetes. Excess retinal de novo lipogenesis-either because of diabetes or because of FAS gain of function-was associated with modestly increased levels of palmitate-containing phosphatidylcholine species in synaptic membranes, a finding with as yet uncertain significance. These findings implicate glucose-dependent increases in photoreceptor de novo lipogenesis in the early pathogenesis of DR, although the mechanism of deleterious action of this pathway remains unclear.


Asunto(s)
Retinopatía Diabética/etiología , Lipogénesis/fisiología , Células Fotorreceptoras de Vertebrados/fisiología , Acetil-CoA Carboxilasa/metabolismo , Animales , Diabetes Mellitus/metabolismo , Retinopatía Diabética/metabolismo , Ácido Graso Sintasas/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Retina/patología
13.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34244440

RESUMEN

Oligodendrocyte precursor cells (OPCs) retain the capacity to remyelinate axons in the corpus callosum (CC) upon demyelination. However, the dynamics of OPC activation, mode of cell division, migration, and differentiation on a single-cell level remain poorly understood due to the lack of longitudinal observations of individual cells within the injured brain. After inducing focal demyelination with lysophosphatidylcholin in the CC of adult mice, we used two-photon microscopy to follow for up to 2 mo OPCs and their differentiating progeny, genetically labeled through conditional recombination driven by the regulatory elements of the gene Achaete-scute homolog 1. OPCs underwent several rounds of symmetric and asymmetric cell divisions, producing a subset of daughter cells that differentiates into myelinating oligodendrocytes. While OPCs continue to proliferate, differentiation into myelinating oligodendrocytes declines with time, and death of OPC-derived daughter cells increases. Thus, chronic in vivo imaging delineates the cellular principles leading to remyelination in the adult brain, providing a framework for the development of strategies to enhance endogenous brain repair in acute and chronic demyelinating disease.


Asunto(s)
Envejecimiento/fisiología , Cuerpo Calloso/diagnóstico por imagen , Imagenología Tridimensional , Remielinización/fisiología , Animales , Muerte Celular , División Celular , Movimiento Celular , Células Clonales , Ratones , Células Precursoras de Oligodendrocitos/citología
14.
Cell Stem Cell ; 28(5): 967-977.e8, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33631115

RESUMEN

Neural stem cells (NSCs) generate neurons throughout life in the hippocampal dentate gyrus. With advancing age, levels of neurogenesis sharply drop, which has been associated with a decline in hippocampal memory function. However, cell-intrinsic mechanisms mediating age-related changes in NSC activity remain largely unknown. Here, we show that the nuclear lamina protein lamin B1 (LB1) is downregulated with age in mouse hippocampal NSCs, whereas protein levels of SUN-domain containing protein 1 (SUN1), previously implicated in Hutchinson-Gilford progeria syndrome (HGPS), increase. Balancing the levels of LB1 and SUN1 in aged NSCs restores the strength of the endoplasmic reticulum diffusion barrier that is associated with segregation of aging factors in proliferating NSCs. Virus-based restoration of LB1 expression in aged NSCs enhances stem cell activity in vitro and increases progenitor cell proliferation and neurogenesis in vivo. Thus, we here identify a mechanism that mediates age-related decline of neurogenesis in the mammalian hippocampus.


Asunto(s)
Envejecimiento , Lamina Tipo B , Células-Madre Neurales , Progeria , Animales , Hipocampo/citología , Ratones , Neurogénesis
15.
Nat Rev Neurosci ; 22(4): 223-236, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33633402

RESUMEN

Neural stem cells (NSCs) generate new neurons throughout life in the mammalian brain. Adult-born neurons shape brain function, and endogenous NSCs could potentially be harnessed for brain repair. In this Review, focused on hippocampal neurogenesis in rodents, we highlight recent advances in the field based on novel technologies (including single-cell RNA sequencing, intravital imaging and functional observation of newborn cells in behaving mice) and characterize the distinct developmental steps from stem cell activation to the integration of newborn neurons into pre-existing circuits. Further, we review current knowledge of how levels of neurogenesis are regulated, discuss findings regarding survival and maturation of adult-born cells and describe how newborn neurons affect brain function. The evidence arguing for (and against) lifelong neurogenesis in the human hippocampus is briefly summarized. Finally, we provide an outlook of what is needed to improve our understanding of the mechanisms and functional consequences of adult neurogenesis and how the field may move towards more translational relevance in the context of acute and chronic neural injury and stem cell-based brain repair.


Asunto(s)
Hipocampo/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Animales , Proliferación Celular/fisiología , Humanos
16.
Curr Opin Cell Biol ; 69: 17-22, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33429112

RESUMEN

Somatic stem cells are required for tissue development, homeostasis, and repair. Recent data suggested that previous biographical experiences of individual stem cells influence their behavior in the context of tissue formation and govern stem cell responses to external stimuli. Here we provide a concise review how a cell's biography, for example, previous rounds of cell divisions or the age-dependent accumulation of cellular damage, is remembered in stem cells and how previous experiences affect the segregation of cellular components, thus guiding cellular behavior in vertebrate stem cells. Further, we suggest future directions of research that may help to unravel the molecular underpinnings of how past experiences guide future cellular behavior.


Asunto(s)
Células Madre , División Celular , Homeostasis
17.
Nat Neurosci ; 24(2): 225-233, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33349709

RESUMEN

Neural stem cells (NSCs) generate neurons throughout life in the mammalian hippocampus. However, the potential for long-term self-renewal of individual NSCs within the adult brain remains unclear. We used two-photon microscopy and followed NSCs that were genetically labeled through conditional recombination driven by the regulatory elements of the stem cell-expressed genes GLI family zinc finger 1 (Gli1) or achaete-scute homolog 1 (Ascl1). Through intravital imaging of NSCs and their progeny, we identify a population of Gli1-targeted NSCs showing long-term self-renewal in the adult hippocampus. In contrast, once activated, Ascl1-targeted NSCs undergo limited proliferative activity before they become exhausted. Using single-cell RNA sequencing, we show that Gli1- and Ascl1-targeted cells have highly similar yet distinct transcriptional profiles, supporting the existence of heterogeneous NSC populations with diverse behavioral properties. Thus, we here identify long-term self-renewing NSCs that contribute to the generation of new neurons in the adult hippocampus.


Asunto(s)
Hipocampo/crecimiento & desarrollo , Células-Madre Neurales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linaje de la Célula , Femenino , Perfilación de la Expresión Génica , Hipocampo/citología , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Microscopía Intravital , Masculino , Metalotioneína 3 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Regeneración Nerviosa , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Análisis de la Célula Individual , Proteína con Dedos de Zinc GLI1/biosíntesis , Proteína con Dedos de Zinc GLI1/genética
18.
STAR Protoc ; 1(2): 100081, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-33000004

RESUMEN

This protocol presents a plate-based workflow to perform RNA sequencing analysis of single cells/nuclei using Smart-seq2. We describe (1) the dissociation procedures for cell/nucleus isolation from the mouse brain and human organoids, (2) the flow sorting of single cells/nuclei into 384-well plates, and (3) the preparation of libraries following miniaturization of the Smart-seq2 protocol using a liquid-handling robot. This pipeline allows for the reliable, high-throughput, and cost-effective preparation of mouse and human samples for full-length deep single-cell/nucleus RNA sequencing. For complete details on the use and execution of this protocol, please refer to Bowers et al. (2020).


Asunto(s)
Análisis de Secuencia de ARN/instrumentación , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos , Animales , Secuencia de Bases/genética , Encéfalo/citología , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Separación Celular/métodos , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Ratones , Miniaturización , ARN/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética , Secuenciación del Exoma/métodos , Flujo de Trabajo
19.
Stem Cell Reports ; 15(3): 566-576, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32857979

RESUMEN

Fatty acid ß-oxidation (FAO), the breakdown of lipids, is a metabolic pathway used by various stem cells. FAO levels are generally high during quiescence and downregulated with proliferation. The endogenous metabolite malonyl-CoA modulates lipid metabolism as a reversible FAO inhibitor and as a substrate for de novo lipogenesis. Here we assessed whether malonyl-CoA can be exploited to steer the behavior of hematopoietic stem/progenitor cells (HSPCs), quiescent stem cells of clinical relevance. Treatment of mouse HSPCs in vitro with malonyl-CoA increases HSPC numbers compared with nontreated controls and ameliorates blood reconstitution capacity when transplanted in vivo, mainly through enhanced lymphoid reconstitution. Similarly, human HSPC numbers also increase upon malonyl-CoA treatment in vitro. These data corroborate that lipid metabolism can be targeted to direct cell fate and stem cell proliferation. Physiological modulation of metabolic pathways, rather than genetic or pharmacological inhibition, provides unique perspectives for stem cell manipulations in health and disease.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Metabolismo de los Lípidos , Linfocitos/citología , Metaboloma , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Células Cultivadas , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Linfocitos/metabolismo , Malonil Coenzima A/metabolismo , Metaboloma/genética , Ratones Endogámicos C57BL , Oxidación-Reducción
20.
Cell Stem Cell ; 27(1): 98-109.e11, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32386572

RESUMEN

Altered neural stem/progenitor cell (NSPC) activity and neurodevelopmental defects are linked to intellectual disability. However, it remains unclear whether altered metabolism, a key regulator of NSPC activity, disrupts human neurogenesis and potentially contributes to cognitive defects. We investigated links between lipid metabolism and cognitive function in mice and human embryonic stem cells (hESCs) expressing mutant fatty acid synthase (FASN; R1819W), a metabolic regulator of rodent NSPC activity recently identified in humans with intellectual disability. Mice homozygous for the FASN R1812W variant have impaired adult hippocampal NSPC activity and cognitive defects because of lipid accumulation in NSPCs and subsequent lipogenic ER stress. Homozygous FASN R1819W hESC-derived NSPCs show reduced rates of proliferation in embryonic 2D cultures and 3D forebrain regionalized organoids, consistent with a developmental phenotype. These data from adult mouse models and in vitro models of human brain development suggest that altered lipid metabolism contributes to intellectual disability.


Asunto(s)
Metabolismo de los Lípidos , Células-Madre Neurales , Animales , Proliferación Celular , Ácido Graso Sintasas , Hipocampo , Trastornos de la Memoria , Ratones , Neurogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA