Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Curr Opin Cell Biol ; 87: 102340, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38401182

RESUMO

Glial fibrillary acidic protein (GFAP) is an intermediate filament (IF) protein expressed in specific types of glial cells in the nervous system. The expression of GFAP is highly regulated during brain development and in neurological diseases. The presence of distinct GFAP-isoforms in various cell types, developmental stages, and diseases indicates that GFAP (post-)transcriptional regulation has a role in glial cell physiology and pathology. GFAP-isoforms differ in sub-cellular localisation, IF-network assembly properties, and IF-dynamics which results in distinct molecular interactions and mechanical properties of the IF-network. Therefore, GFAP (post-)transcriptional regulation is likely a mechanism by which radial glia, astrocytes, and glioma cells can modulate cellular function.


Assuntos
Astrócitos , Filamentos Intermediários , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Filamentos Intermediários/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Isoformas de Proteínas/genética , Regulação da Expressão Gênica
2.
Mikrochim Acta ; 191(1): 71, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38168828

RESUMO

The complex structure and function of the human central nervous system that develops from the neural tube made in vitro modeling quite challenging until the discovery of brain organoids. Human-induced pluripotent stem cells-derived brain organoids offer recapitulation of the features of early human neurodevelopment in vitro, including the generation, proliferation, and differentiation into mature neurons and micro-macroglial cells, as well as the complex interactions among these diverse cell types of the developing brain. Recent advancements in brain organoids, microfluidic systems, real-time sensing technologies, and their cutting-edge integrated use provide excellent models and tools for emulation of fundamental neurodevelopmental processes, the pathology of neurological disorders, personalized transplantation therapy, and high-throughput neurotoxicity testing by bridging the gap between two-dimensional models and the complex three-dimensional environment in vivo. In this review, we summarize how bioengineering approaches are applied to mitigate the limitations of brain organoids for biomedical and clinical research. We further provide an extensive overview and future perspectives of the humanized brain organoids-on-chip platforms with integrated sensors toward brain organoid intelligence and biocomputing studies. Such approaches might pave the way for increasing approvable clinical applications by solving their current limitations.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças do Sistema Nervoso , Humanos , Encéfalo , Neurônios , Células-Tronco Pluripotentes Induzidas/metabolismo , Organoides
3.
Cell Rep ; 40(1): 111029, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35793630

RESUMO

The habenula plays a key role in various motivated and pathological behaviors and is composed of molecularly distinct neuron subtypes. Despite progress in identifying mature habenula neuron subtypes, how these subtypes develop and organize into functional brain circuits remains largely unknown. Here, we performed single-cell transcriptional profiling of mouse habenular neurons at critical developmental stages, instructed by detailed three-dimensional anatomical data. Our data reveal cellular and molecular trajectories during embryonic and postnatal development, leading to different habenular subtypes. Further, based on this analysis, our work establishes the distinctive functional properties and projection target of a subtype of Cartpt+ habenula neurons. Finally, we show how comparison of single-cell transcriptional profiles and GWAS data links specific developing habenular subtypes to psychiatric disease. Together, our study begins to dissect the mechanisms underlying habenula neuron subtype-specific development and creates a framework for further interrogation of habenular development in normal and disease states.


Assuntos
Habenula , Animais , Habenula/fisiologia , Camundongos , Neurogênese/genética , Neurônios
4.
EMBO J ; 41(11): e110409, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35451150

RESUMO

Astrocytes are highly abundant in the mammalian brain, and their functions are of vital importance for all aspects of development, adaption, and aging of the central nervous system (CNS). Mounting evidence indicates the important contributions of astrocytes to a wide range of neuropathies. Still, our understanding of astrocyte development significantly lags behind that of other CNS cells. We here combine immunohistochemical approaches with genetic fate-mapping, behavioural paradigms, single-cell transcriptomics, and in vivo two-photon imaging, to comprehensively assess the generation and the proliferation of astrocytes in the dentate gyrus (DG) across the life span of a mouse. Astrogenesis in the DG is initiated by radial glia-like neural stem cells giving rise to locally dividing astrocytes that enlarge the astrocyte compartment in an outside-in-pattern. Also in the adult DG, the vast majority of astrogenesis is mediated through the proliferation of local astrocytes. Interestingly, locally dividing astrocytes were able to adapt their proliferation to environmental and behavioral stimuli revealing an unexpected plasticity. Our study establishes astrocytes as enduring plastic elements in DG circuits, implicating a vital contribution of astrocyte dynamics to hippocampal plasticity.


Assuntos
Células-Tronco Neurais , Neurogênese , Animais , Astrócitos/fisiologia , Giro Denteado , Hipocampo/fisiologia , Mamíferos , Camundongos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia
5.
Cell Rep ; 38(9): 110440, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235796

RESUMO

Spinal cord ependymal cells display neural stem cell properties in vitro and generate scar-forming astrocytes and remyelinating oligodendrocytes after injury. We report that ependymal cells are functionally heterogeneous and identify a small subpopulation (8% of ependymal cells and 0.1% of all cells in a spinal cord segment), which we denote ependymal A (EpA) cells, that accounts for the in vitro stem cell potential in the adult spinal cord. After spinal cord injury, EpA cells undergo self-renewing cell division as they give rise to differentiated progeny. Single-cell transcriptome analysis revealed a loss of ependymal cell gene expression programs as EpA cells gained signaling entropy and dedifferentiated to a stem-cell-like transcriptional state after an injury. We conclude that EpA cells are highly differentiated cells that can revert to a stem cell state and constitute a therapeutic target for spinal cord repair.


Assuntos
Células-Tronco Neurais , Traumatismos da Medula Espinal , Diferenciação Celular/fisiologia , Humanos , Células-Tronco Neurais/metabolismo , Neuroglia , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo
6.
Nat Commun ; 13(1): 1036, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210419

RESUMO

Following the decline of neurogenesis at birth, progenitors of the subventricular zone (SVZ) remain mostly in a quiescent state in the adult human brain. The mechanisms that regulate this quiescent state are still unclear. Here, we isolate CD271+ progenitors from the aged human SVZ for single-cell RNA sequencing analysis. Our transcriptome data reveal the identity of progenitors of the aged human SVZ as late oligodendrocyte progenitor cells. We identify the Wnt pathway antagonist SFRP1 as a possible signal that promotes quiescence of progenitors from the aged human SVZ. Administration of WAY-316606, a small molecule that inhibits SFRP1 function, stimulates activation of neural stem cells both in vitro and in vivo under homeostatic conditions. Our data unravel a possible mechanism through which progenitors of the adult human SVZ are maintained in a quiescent state and a potential target for stimulating progenitors to re-activate.


Assuntos
Ventrículos Laterais , Células-Tronco Neurais , Idoso , Encéfalo/metabolismo , Diferenciação Celular/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ventrículos Laterais/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Transcriptoma
7.
Stem Cell Reports ; 13(4): 627-641, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31522975

RESUMO

Organoid technologies have become a powerful emerging tool to model liver diseases, for drug screening, and for personalized treatments. These applications are, however, limited in their capacity to generate functional hepatocytes in a reproducible and efficient manner. Here, we generated and characterized the hepatic organoid (eHEPO) culture system using human induced pluripotent stem cell (iPSC)-derived EpCAM-positive endodermal cells as an intermediate. eHEPOs can be produced within 2 weeks and expanded long term (>16 months) without any loss of differentiation capacity to mature hepatocytes. Starting from patient-specific iPSCs, we modeled citrullinemia type 1, a urea cycle disorder caused by mutations in the argininosuccinate synthetase (ASS1) enzyme. The disease-related ammonia accumulation phenotype in eHEPOs could be reversed by the overexpression of the wild-type ASS1 gene, which also indicated that this model is amenable to genetic manipulation. Thus, eHEPOs are excellent unlimited cell sources to generate functional hepatic organoids in a fast and efficient manner.


Assuntos
Diferenciação Celular , Suscetibilidade a Doenças , Endoderma/citologia , Hepatócitos/citologia , Fígado/citologia , Fígado/embriologia , Organogênese , Organoides/citologia , Biomarcadores , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Técnicas de Cultura de Tecidos
8.
Cell Stem Cell ; 25(3): 342-356.e7, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31422913

RESUMO

The gastric corpus epithelium is the thickest part of the gastrointestinal tract and is rapidly turned over. Several markers have been proposed for gastric corpus stem cells in both isthmus and base regions. However, the identity of isthmus stem cells (IsthSCs) and the interaction between distinct stem cell populations is still under debate. Here, based on unbiased genetic labeling and biophysical modeling, we show that corpus glands are compartmentalized into two independent zones, with slow-cycling stem cells maintaining the base and actively cycling stem cells maintaining the pit-isthmus-neck region through a process of "punctuated" neutral drift dynamics. Independent lineage tracing based on Stmn1 and Ki67 expression confirmed that rapidly cycling IsthSCs maintain the pit-isthmus-neck region. Finally, single-cell RNA sequencing (RNA-seq) analysis is used to define the molecular identity and lineage relationship of a single, cycling, IsthSC population. These observations define the identity and functional behavior of IsthSCs.


Assuntos
Células-Tronco Adultas/citologia , Mucosa Gástrica/citologia , Estômago/citologia , Células-Tronco Adultas/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Células Cultivadas , Mucosa Gástrica/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Estatmina/metabolismo , Nicho de Células-Tronco
9.
Acta Neuropathol Commun ; 7(1): 84, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31159890

RESUMO

It is currently accepted that the human brain has a limited neurogenic capacity and an impaired regenerative potential. We have previously shown the existence of CD271-expressing neural stem cells (NSCs) in the subventricular zone (SVZ) of Parkinson's disease (PD) patients, which proliferate and differentiate towards neurons and glial cells in vitro. To study the molecular profile of these NSCs in detail, we performed RNA sequencing and mass spectrometry on CD271+ NSCs isolated from human post-mortem SVZ and on homogenates of the SVZ. CD271+ cells were isolated through magnetic cell separation (MACS). We first compared the molecular profile of CD271+ NSCs to the SVZ homogenate from control donors and then compared CD271+ cells to CD11b+ microglia. These results confirmed their neural stem cell identity. Finally we compared controls and PD patients to establish a specific molecular profile of NSCs and the SVZ in PD. While our transcriptome analysis did not identify any differentially expressed genes in the SVZ between control and PD patients, our proteome analysis revealed several proteins that were differentially expressed in PD. Some of these proteins are involved in cytoskeletal organization and mitochondrial function. Transcriptome and proteome analyses of NSCs from PD revealed changes in the expression of genes and proteins involved in metabolism, transcriptional activity and cytoskeletal organization. Our data suggest that NSCs may transit into a primed-quiescent state, that is in an "alert" non-proliferative phase in PD. Our results not only confirm pathological hallmarks of PD (e.g. impaired mitochondrial function), but also show that the NSCs from SVZ undergo significant changes at both transcriptome and proteome level following PD.


Assuntos
Ventrículos Laterais/metabolismo , Células-Tronco Neurais/metabolismo , Doença de Parkinson/metabolismo , Proteoma , Transcriptoma , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(29): 14630-14638, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31253707

RESUMO

Mammalian epidermal stem cells maintain homeostasis of the skin epidermis and contribute to its regeneration throughout adult life. While 2D mouse epidermal stem cell cultures have been established decades ago, a long-term, feeder cell- and serum-free culture system recapitulating murine epidermal architecture has not been available. Here we describe an epidermal organoid culture system that allows long-term, genetically stable expansion of adult epidermal stem cells. Our epidermal expansion media combines atypically high calcium concentrations, activation of cAMP, FGF, and R-spondin signaling with inhibition of bone morphogenetic protein (BMP) signaling. Organoids are established robustly from adult mouse skin and expand over at least 6 mo, while maintaining the basal-apical organization of the mouse interfollicular epidermis. The system represents a powerful tool to study epidermal homeostasis and disease in vitro.


Assuntos
Células-Tronco Adultas/fisiologia , Técnicas de Cultura de Células/métodos , Epiderme/fisiologia , Queratinócitos/fisiologia , Organoides/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Introdução de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética , Fatores de Tempo
11.
Proc Natl Acad Sci U S A ; 115(52): E12245-E12254, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30530645

RESUMO

The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By single-cell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adult myocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury.


Assuntos
Proliferação de Células , Traumatismos Cardíacos/fisiopatologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Traumatismos Cardíacos/genética , Traumatismos Cardíacos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Gravidez , Células-Tronco/citologia , Células-Tronco/metabolismo
12.
Cell ; 175(6): 1591-1606.e19, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30500538

RESUMO

The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes.


Assuntos
Proliferação de Células , Hepatócitos/metabolismo , Organoides/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Hepatócitos/citologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Organoides/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo
13.
Vet Dermatol ; 29(5): 375-e126, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29963730

RESUMO

BACKGROUND: Perturbations of epidermal and follicular homeostasis have been attributed to a variety of skin diseases affecting dogs. The availability of an in vitro system to investigate these diseases is important to understand underlying pathomechanisms. OBJECTIVES: To establish an accurate and reliable in vitro 3D system of canine keratinocyte organoids to lay the basis for studying functional defects in interfollicular epidermis (IFE) and hair follicle (HF) morphogenesis, reconstitution and differentiation that lead to alopecic and epidermal diseases. ANIMALS: Skin biopsies were obtained from freshly euthanized dogs of different breeds with no skin abnormalities. METHODS: Cells derived from microdissected IFE and HFs were seeded in Matrigel and keratinocyte organoids were grown and characterized using immunohistochemistry, RT-qPCR and RNA sequencing. RESULTS: Both organoid lines develop into a basal IFE-like cell type. Gene and protein expression analysis revealed high mRNA and protein levels of keratins 5 and 14, IFE differentiation markers and intercellular molecules. Key markers of HF stem cells were lacking. Withdrawal of growth factors resulted in upregulation of markers such as KRT16, Involucrin, KRT17 and SOX9, showing the potential of the organoids to develop towards more differentiated tissue. CONCLUSION AND CLINICAL IMPORTANCE: Our 3D in vitro culture system provides the basis to explore epidermal function, to investigate the culture conditions necessary for the development of organoids with a HF signature and to address cutaneous disorders in dogs. However, for induction of HF signatures or hair growth, addition of different growth factors or co-culture with dermal papilla will be required.


Assuntos
Cães/anatomia & histologia , Queratinócitos/citologia , Técnicas de Cultura de Órgãos/veterinária , Animais , Biópsia/veterinária , Células Cultivadas/citologia , Células Epidérmicas , Queratinócitos/patologia , Técnicas de Cultura de Órgãos/métodos , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Análise de Sequência de RNA/veterinária , Pele/citologia , Pele/patologia , Dermatopatias/patologia , Dermatopatias/veterinária
14.
Nat Commun ; 9(1): 1898, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29765026

RESUMO

Hematopoietic stem cells (HSCs) continuously replenish all blood cell types through a series of differentiation steps and repeated cell divisions that involve the generation of lineage-committed progenitors. However, whether cell division in HSCs precedes differentiation is unclear. To this end, we used an HSC cell-tracing approach and Ki67RFP knock-in mice, in a non-conditioned transplantation model, to assess divisional history, cell cycle progression, and differentiation of adult HSCs. Our results reveal that HSCs are able to differentiate into restricted progenitors, especially common myeloid, megakaryocyte-erythroid and pre-megakaryocyte progenitors, without undergoing cell division and even before entering the S phase of the cell cycle. Additionally, the phenotype of the undivided but differentiated progenitors correlated with the expression of lineage-specific genes and loss of multipotency. Thus HSC fate decisions can be uncoupled from physical cell division. These results facilitate a better understanding of the mechanisms that control fate decisions in hematopoietic cells.


Assuntos
Divisão Celular , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células Progenitoras Mieloides/citologia , Animais , Feminino , Citometria de Fluxo , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL
15.
Proc Natl Acad Sci U S A ; 115(4): E610-E619, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29311336

RESUMO

The adult mouse subependymal zone provides a niche for mammalian neural stem cells (NSCs). However, the molecular signature, self-renewal potential, and fate behavior of NSCs remain poorly defined. Here we propose a model in which the fate of active NSCs is coupled to the total number of neighboring NSCs in a shared niche. Using knock-in reporter alleles and single-cell RNA sequencing, we show that the Wnt target Tnfrsf19/Troy identifies both active and quiescent NSCs. Quantitative analysis of genetic lineage tracing of individual NSCs under homeostasis or in response to injury reveals rapid expansion of stem-cell number before some return to quiescence. This behavior is best explained by stochastic fate decisions, where stem-cell number within a shared niche fluctuates over time. Fate mapping proliferating cells using a Ki67iresCreER allele confirms that active NSCs reversibly return to quiescence, achieving long-term self-renewal. Our findings suggest a niche-based mechanism for the regulation of NSC fate and number.


Assuntos
Ventrículos Laterais/citologia , Células-Tronco Neurais/fisiologia , Nicho de Células-Tronco , Animais , Linhagem da Célula , Proliferação de Células , Camundongos , Neurogênese , Receptores do Fator de Necrose Tumoral/metabolismo , Análise de Célula Única , Transcriptoma
16.
Stem Cell Reports ; 9(5): 1530-1545, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29141232

RESUMO

Mutations in FOXP1 have been linked to neurodevelopmental disorders including intellectual disability and autism; however, the underlying molecular mechanisms remain ill-defined. Here, we demonstrate with RNA and chromatin immunoprecipitation sequencing that FOXP1 directly regulates genes controlling neurogenesis. We show that FOXP1 is expressed in embryonic neural stem cells (NSCs), and modulation of FOXP1 expression affects both neuron and astrocyte differentiation. Using a murine model of cortical development, FOXP1-knockdown in utero was found to reduce NSC differentiation and migration during corticogenesis. Furthermore, transplantation of FOXP1-knockdown NSCs in neonatal mice after hypoxia-ischemia challenge demonstrated that FOXP1 is also required for neuronal differentiation and functionality in vivo. FOXP1 was found to repress the expression of Notch pathway genes including the Notch-ligand Jagged1, resulting in inhibition of Notch signaling. Finally, blockade of Jagged1 in FOXP1-knockdown NSCs rescued neuronal differentiation in vitro. Together, these data support a role for FOXP1 in regulating embryonic NSC differentiation by modulating Notch signaling.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Neurais/citologia , Neurogênese , Proteínas Repressoras/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Fatores de Transcrição Forkhead/genética , Hipóxia-Isquemia Encefálica/terapia , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Repressoras/genética , Transplante de Células-Tronco
17.
Stem Cell Reports ; 8(6): 1472-1478, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28506535

RESUMO

Blood cell generation depends on continuous cellular output by the sequential hierarchy of hematopoietic stem cell (HSC) and progenitor populations that all contain quiescent and actively cycling cells. Hematopoietic stem and progenitor cells (HSPCs) express the surface molecule Stem cell antigen 1 (SCA-1/LY6A). Using histone 2B-red fluorescent fusion protein label retention and cell-cycle reporter mice, we demonstrate that high SCA-1 expression (SCA-1hi) identifies not only quiescent HSCs but quiescent cells on all hierarchical levels within the lineage-SCA-1+KIT+ (LSK) population. Each transplanted SCA-1hi HSPC population also displayed self-renewal potential superior to that of the respective SCA-1lo population. SCA-1 expression is inducible by type I interferon (IFN). We show, however, that quiescence and high self-renewal capacity of cells with brighter SCA-1 expression at steady state were independent of type I IFN signaling. We conclude that SCA-1 expression levels can be used to prospectively isolate functionally heterogeneous HSPC subpopulations.


Assuntos
Antígenos Ly/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Membrana/metabolismo , Animais , Antígenos Ly/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Autorrenovação Celular , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Histonas/genética , Histonas/metabolismo , Interferon Tipo I/metabolismo , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais , Transplante Homólogo
19.
Cell Stem Cell ; 20(2): 177-190.e4, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-27939219

RESUMO

Lgr5+ adult intestinal stem cells are highly proliferative throughout life. Single Lgr5+ stem cells can be cultured into three-dimensional organoids containing all intestinal epithelial cell types at near-normal ratios. Conditions to generate the main cell types (enterocyte, goblet cells, Paneth cells, and M cells) are well established, but signals to induce the spectrum of hormone-producing enteroendocrine cells (EECs) have remained elusive. Here, we induce Lgr5+ stem cell quiescence in vitro by blocking epidermal growth factor receptor (EGFR) or mitogen-associated protein kinase (MAPK) signaling pathways in organoids and show that their quiescent state is readily reverted. Quiescent Lgr5+ stem cells acquire a distinct molecular signature biased toward EEC differentiation. Indeed, combined inhibition of Wnt, Notch, and MAPK pathways efficiently generates a diversity of EEC hormone-expressing subtypes in vitro. Our observations uncouple Wnt-dependent stem cell maintenance from EGF-dependent proliferation and provide an approach for the study of the elusive EECs in a defined environment.


Assuntos
Ciclo Celular , Diferenciação Celular , Células Enteroendócrinas/citologia , Hormônios/biossíntese , Intestinos/citologia , Organoides/citologia , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/citologia , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Perfilação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Receptores Notch/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única , Proteínas Wnt/metabolismo
20.
Nat Immunol ; 17(12): 1467-1478, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27776108

RESUMO

Tissue-resident memory T cells (TRM cells) in the airways mediate protection against respiratory infection. We characterized TRM cells expressing integrin αE (CD103) that reside within the epithelial barrier of human lungs. These cells had specialized profiles of chemokine receptors and adhesion molecules, consistent with their unique localization. Lung TRM cells were poised for rapid responsiveness by constitutive expression of deployment-ready mRNA encoding effector molecules, but they also expressed many inhibitory regulators, suggestive of programmed restraint. A distinct set of transcription factors was active in CD103+ TRM cells, including Notch. Genetic and pharmacological experiments with mice revealed that Notch activity was required for the maintenance of CD103+ TRM cells. We have thus identified specialized programs underlying the residence, persistence, vigilance and tight control of human lung TRM cells.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Memória Imunológica , Vírus da Influenza A Subtipo H3N2/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Infecções Respiratórias/imunologia , Animais , Antígenos CD/metabolismo , Células Cultivadas , Feminino , Humanos , Cadeias alfa de Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Receptor Notch1/genética , Receptor Notch2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA