Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Commun Biol ; 6(1): 996, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773433

RESUMEN

Protection of telomeres 1a (POT1a) is a telomere binding protein. A decrease of POT1a is related to myeloid-skewed haematopoiesis with ageing, suggesting that protection of telomeres is essential to sustain multi-potency. Since mesenchymal stem cells (MSCs) are a constituent of the hematopoietic niche in bone marrow, their dysfunction is associated with haematopoietic failure. However, the importance of telomere protection in MSCs has yet to be elucidated. Here, we show that genetic deletion of POT1a in MSCs leads to intracellular accumulation of fatty acids and excessive ROS and DNA damage, resulting in impaired osteogenic-differentiation. Furthermore, MSC-specific POT1a deficient mice exhibited skeletal retardation due to reduction of IL-7 producing bone lining osteoblasts. Single-cell gene expression profiling of bone marrow from POT1a deficient mice revealed that B-lymphopoiesis was selectively impaired. These results demonstrate that bone marrow microenvironments composed of POT1a deficient MSCs fail to support B-lymphopoiesis, which may underpin age-related myeloid-bias in haematopoiesis.


Asunto(s)
Linfopoyesis , Telómero , Animales , Ratones , Envejecimiento , Diferenciación Celular , Linfopoyesis/genética , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
2.
Blood ; 142(19): 1622-1632, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37562000

RESUMEN

A critical regulatory role of hematopoietic stem cell (HSC) vascular niches in the bone marrow has been implicated to occur through endothelial niche cell expression of KIT ligand. However, endothelial-derived KIT ligand is expressed in both a soluble and membrane-bound form and not unique to bone marrow niches, and it is also systemically distributed through the circulatory system. Here, we confirm that upon deletion of both the soluble and membrane-bound forms of endothelial-derived KIT ligand, HSCs are reduced in mouse bone marrow. However, the deletion of endothelial-derived KIT ligand was also accompanied by reduced soluble KIT ligand levels in the blood, precluding any conclusion as to whether the reduction in HSC numbers reflects reduced endothelial expression of KIT ligand within HSC niches, elsewhere in the bone marrow, and/or systemic soluble KIT ligand produced by endothelial cells outside of the bone marrow. Notably, endothelial deletion, specifically of the membrane-bound form of KIT ligand, also reduced systemic levels of soluble KIT ligand, although with no effect on stem cell numbers, implicating an HSC regulatory role primarily of soluble rather than membrane KIT ligand expression in endothelial cells. In support of a role of systemic rather than local niche expression of soluble KIT ligand, HSCs were unaffected in KIT ligand deleted bones implanted into mice with normal systemic levels of soluble KIT ligand. Our findings highlight the need for more specific tools to unravel niche-specific roles of regulatory cues expressed in hematopoietic niche cells in the bone marrow.


Asunto(s)
Células Endoteliales , Factor de Células Madre , Ratones , Animales , Factor de Células Madre/metabolismo , Células Madre Hematopoyéticas/metabolismo , Médula Ósea/metabolismo , Huesos , Nicho de Células Madre , Células de la Médula Ósea/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(32): e2206860120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523546

RESUMEN

Mbtd1 (mbt domain containing 1) encodes a nuclear protein containing a zinc finger domain and four malignant brain tumor (MBT) repeats. We previously generated Mbtd1-deficient mice and found that MBTD1 is highly expressed in fetal hematopoietic stem cells (HSCs) and sustains the number and function of fetal HSCs. However, since Mbtd1-deficient mice die soon after birth possibly due to skeletal abnormalities, its role in adult hematopoiesis remains unclear. To address this issue, we generated Mbtd1 conditional knockout mice and analyzed adult hematopoietic tissues deficient in Mbtd1. We observed that the numbers of HSCs and progenitors increased and Mbtd1-deficient HSCs exhibited hyperactive cell cycle, resulting in a defective response to exogenous stresses. Mechanistically, we found that MBTD1 directly binds to the promoter region of FoxO3a, encoding a forkhead protein essential for HSC quiescence, and interacts with components of TIP60 chromatin remodeling complex and other proteins involved in HSC and other stem cell functions. Restoration of FOXO3a activity in Mbtd1-deficient HSCs in vivo rescued cell cycle and pool size abnormalities. These findings indicate that MBTD1 is a critical regulator for HSC pool size and function, mainly through the maintenance of cell cycle quiescence by FOXO3a.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Ratones , Ciclo Celular/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/metabolismo
4.
Elife ; 122023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37266576

RESUMEN

Polycomb repressive complex (PRC) 1 regulates stem cell fate by mediating mono-ubiquitination of histone H2A at lysine 119. While canonical PRC1 is critical for hematopoietic stem and progenitor cell (HSPC) maintenance, the role of non-canonical PRC1 in hematopoiesis remains elusive. PRC1.1, a non-canonical PRC1, consists of PCGF1, RING1B, KDM2B, and BCOR. We recently showed that PRC1.1 insufficiency induced by the loss of PCGF1 or BCOR causes myeloid-biased hematopoiesis and promotes transformation of hematopoietic cells in mice. Here we show that PRC1.1 serves as an epigenetic switch that coordinates homeostatic and emergency hematopoiesis. PRC1.1 maintains balanced output of steady-state hematopoiesis by restricting C/EBPα-dependent precocious myeloid differentiation of HSPCs and the HOXA9- and ß-catenin-driven self-renewing network in myeloid progenitors. Upon regeneration, PRC1.1 is transiently inhibited to facilitate formation of granulocyte-macrophage progenitor (GMP) clusters, thereby promoting emergency myelopoiesis. Moreover, constitutive inactivation of PRC1.1 results in unchecked expansion of GMPs and eventual transformation. Collectively, our results define PRC1.1 as a novel critical regulator of emergency myelopoiesis, dysregulation of which leads to myeloid transformation.


Asunto(s)
Mielopoyesis , Complejo Represivo Polycomb 1 , Animales , Ratones , Complejo Represivo Polycomb 1/metabolismo , Mielopoyesis/genética , Histonas , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo
5.
Nature ; 615(7954): 900-906, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922585

RESUMEN

Sex chromosome disorders severely compromise gametogenesis in both males and females. In oogenesis, the presence of an additional Y chromosome or the loss of an X chromosome disturbs the robust production of oocytes1-5. Here we efficiently converted the XY chromosome set to XX without an additional Y chromosome in mouse pluripotent stem (PS) cells. In addition, this chromosomal alteration successfully eradicated trisomy 16, a model of Down's syndrome, in PS cells. Artificially produced euploid XX PS cells differentiated into mature oocytes in culture with similar efficiency to native XX PS cells. Using this method, we differentiated induced pluripotent stem cells from the tail of a sexually mature male mouse into fully potent oocytes, which gave rise to offspring after fertilization. This study provides insights that could ameliorate infertility caused by sex chromosome or autosomal disorders, and opens the possibility of bipaternal reproduction.


Asunto(s)
Ingeniería Genética , Técnicas In Vitro , Oocitos , Cromosoma X , Animales , Femenino , Masculino , Ratones , Oocitos/metabolismo , Oocitos/fisiología , Cromosoma X/genética , Cromosoma Y/genética , Células Madre Pluripotentes/metabolismo , Síndrome de Down/genética , Síndrome de Down/terapia , Fertilización , Infertilidad/terapia , Homosexualidad Masculina , Trastornos de los Cromosomas Sexuales/complicaciones , Trastornos de los Cromosomas Sexuales/genética , Trastornos de los Cromosomas Sexuales/terapia , Ingeniería Genética/métodos
6.
Methods Mol Biol ; 2520: 171-180, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35575971

RESUMEN

The zebrafish is a useful model to identify genes functioning in hematopoiesis, owing to high conservation of hematopoiesis. Flow cytometry is widely used to isolate and quantitatively characterize human and mouse hematopoietic cells, often using fluorescently labeled antibodies. However, such analysis is limited in zebrafish due to lack of antibodies that recognize zebrafish hematopoietic cells. We here describe methods for isolation of hematopoietic cells by antibody-free flow cytometry in the zebrafish embryo. Hematopoietic stem cells (HSCs) are specified from a specific subset of vascular endothelial cells, the hemogenic endothelial cell (HEC), by a high level of Notch signaling. In combination with a Notch reporter line (Tp1:GFP) and a vascular specific line (fli1a:dsRed), HECs can be isolated as Tp1:GFPhigh fli1a:dsRed+ cells at 20-22 hours post-fertilization (hpf). Zebrafish erythrocytes can be purified using 1,5-bis{[2-(dimethylamino)ethyl]amino}-4, 8-dihydroxyanthracene-9,10-dione (DRAQ5), a DNA-staining fluorescent dye, and gata1:dsRed (erythroid marker line). DRAQ5high dsRed+ cells are morphologically erythrocyte-like, hemoglobin-stained positive, and express erythropoiesis-related genes. Zebrafish neutrophils can be also isolated using the lectin Phaseolus vulgaris erythroagglutinin (PHA-E) and DRAQ5. PHA-Elow DRAQ5low cells have myeloperoxidase activity, are Sudan Black B-positive, and express neutrophil-related genes. These methods will help to perform genetical and functional assays for various types of hematopoietic cells in zebrafish embryos.


Asunto(s)
Citometría de Flujo , Células Madre Hematopoyéticas , Pez Cebra , Animales , Separación Celular/métodos , Embrión no Mamífero , Células Endoteliales , Eritropoyesis , Citometría de Flujo/métodos , Hematopoyesis , Proteínas de Pez Cebra
7.
Cell Rep ; 39(6): 110805, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545056

RESUMEN

Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells (HSCs), characterized by ineffective hematopoiesis and frequent progression to leukemia. It has long remained unresolved how MDS cells, which are less proliferative, inhibit normal hematopoiesis and eventually dominate the bone marrow space. Despite several studies implicating mesenchymal stromal or stem cells (MSCs), a principal component of the HSC niche, in the inhibition of normal hematopoiesis, the molecular mechanisms underlying this process remain unclear. Here, we demonstrate that both human and mouse MDS cells perturb bone metabolism by suppressing the osteolineage differentiation of MSCs, which impairs the ability of MSCs to support normal HSCs. Enforced MSC differentiation rescues the suppressed normal hematopoiesis in both in vivo and in vitro MDS models. Intriguingly, the suppression effect is reversible and mediated by extracellular vesicles (EVs) derived from MDS cells. These findings shed light on the novel MDS EV-MSC axis in ineffective hematopoiesis.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Síndromes Mielodisplásicos , Animales , Vesículas Extracelulares/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Síndromes Mielodisplásicos/metabolismo
8.
Cell Stem Cell ; 28(2): 187-190, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33545078

RESUMEN

Epigenetic memories play an important part in regulating stem cell identities. Tools from the theory of non-Markov processes may help us understand these memories better and develop a more integrated view of stem cell fate and function.


Asunto(s)
Epigénesis Genética , Células Madre , Diferenciación Celular
9.
Commun Biol ; 3(1): 736, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277618

RESUMEN

Biomedical research often involves conducting experiments on model organisms in the anticipation that the biology learnt will transfer to humans. Previous comparative studies of mouse and human tissues were limited by the use of bulk-cell material. Here we show that transfer learning-the branch of machine learning that concerns passing information from one domain to another-can be used to efficiently map bone marrow biology between species, using data obtained from single-cell RNA sequencing. We first trained a multiclass logistic regression model to recognize different cell types in mouse bone marrow achieving equivalent performance to more complex artificial neural networks. Furthermore, it was able to identify individual human bone marrow cells with 83% overall accuracy. However, some human cell types were not easily identified, indicating important differences in biology. When re-training the mouse classifier using data from human, less than 10 human cells of a given type were needed to accurately learn its representation. In some cases, human cell identities could be inferred directly from the mouse classifier via zero-shot learning. These results show how simple machine learning models can be used to reconstruct complex biology from limited data, with broad implications for biomedical research.


Asunto(s)
Células de la Médula Ósea/clasificación , Aprendizaje Automático , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Animales , Separación Celular , Humanos , Ratones
10.
Cell Syst ; 11(6): 640-652.e5, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33296684

RESUMEN

Changes in stem cell activity may underpin aging. However, these changes are not completely understood. Here, we combined single-cell profiling with machine learning and in vivo functional studies to explore how hematopoietic stem cell (HSC) divisions patterns evolve with age. We first trained an artificial neural network (ANN) to accurately identify cell types in the hematopoietic hierarchy and predict their age from single-cell gene-expression patterns. We then used this ANN to compare identities of daughter cells immediately after HSC divisions and found that the self-renewal ability of individual HSCs declines with age. Furthermore, while HSC cell divisions are deterministic and intrinsically regulated in young and old age, they are variable and niche sensitive in mid-life. These results indicate that the balance between intrinsic and extrinsic regulation of stem cell activity alters substantially with age and help explain why stem cell numbers increase through life, yet regenerative potency declines.


Asunto(s)
Envejecimiento/inmunología , Diferenciación Celular/inmunología , División Celular/inmunología , Aprendizaje Automático/normas , Células Madre Hematopoyéticas/metabolismo , Humanos
11.
Phys Biol ; 17(6): 065013, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33210617

RESUMEN

Modern single cell experiments have revealed unexpected heterogeneity in apparently functionally 'pure' cell populations. However, we are still lacking a conceptual framework to understand this heterogeneity. Here, we propose that cellular memories-changes in the molecular status of a cell in response to a stimulus, that modify the ability of the cell to respond to future stimuli-are an essential ingredient in any such theory. We illustrate this idea by considering a simple age-structured model of stem cell proliferation that takes account of mitotic memories. Using this model we argue that asynchronous mitosis generates heterogeneity that is central to stem cell population function. This model naturally explains why stem cell numbers increase through life, yet regenerative potency simultaneously declines.


Asunto(s)
Mitosis , Células Madre/fisiología , Modelos Biológicos
12.
Rinsho Ketsueki ; 61(9): 1064-1070, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-33162500

RESUMEN

Expansion of stem cell numbers without reduction in their regenerative potential is crucial for therapeutic applications. However, the repeated cell divisions and aging impair stem cell function. We found that Pot1a, a component of the shelterin that protects telomeres, involves the maintenance of hematopoietic stem cell (HSC) activity during aging. Pot1a maintained the self-renewal activity of HSCs through the prevention of DNA damage responses in HSCs and suppression of the production of reactive oxygen species. Furthermore, the exogenous Pot1a expanded the HSC number and rejuvenated aged HSCs function upon ex vivo culture. Consistent with these results, treatment with exogenous human POT1 protein maintains human HSC activity in culture. Collectively, these results show that Pot1a or POT1 sustains HSC activity and can be used to expand HSC numbers ex vivo.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Envejecimiento , División Celular , Humanos , Mantenimiento , Complejo Shelterina , Telómero , Proteínas de Unión a Telómeros
13.
Genes Cells ; 25(12): 770-781, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33006802

RESUMEN

Zebrafish is a useful model to study vertebrate hematopoiesis, but lack of antibodies to zebrafish proteins has limited purification of hematopoietic cells. Here, we purified neutrophils from larval and adult zebrafish using the lectin Phaseolus vulgaris erythroagglutinin (PHA-E) and DRAQ5, a DNA-staining fluorescent dye. In adult kidney marrow, we purified neutrophil-like PHA-E4low DRAQ5low cells, which neutrophil-type granules. Specifically, at 96-hr post-fertilization, we sorted large-sized cells from larvae using forward scatter and found that they consisted of PHA-Elow DRAQ5low populations. These cells had myeloperoxidase activity, were Sudan Black B-positive and expressed high levels of neutrophil-specific (csf3r and mpx) mRNAs, all neutrophil characteristics. Using this method, we conducted functional analysis suggesting that zyxin (Zyx) plays a role in neutrophil generation in zebrafish larvae. Overall, PHA-E and DRAQ5-based flow cytometry serves as a tool to purify zebrafish neutrophils.


Asunto(s)
Citometría de Flujo/métodos , Hematopoyesis , Neutrófilos/citología , Cultivo Primario de Células/métodos , Animales , Células Cultivadas , Lectinas/metabolismo , Neutrófilos/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
14.
iScience ; 23(11): 101654, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33103089

RESUMEN

p32/C1qbp regulates mitochondrial protein synthesis and is essential for oxidative phosphorylation in mitochondria. Although dysfunction of p32/C1qbp impairs fetal development and immune responses, its role in hematopoietic differentiation remains unclear. Here, we found that mitochondrial dysfunction affected terminal differentiation of newly identified erythroid/B-lymphoid progenitors among CD45- Ter119- CD31- triple-negative cells (TNCs) in bone marrow. Hematopoietic cell-specific genetic deletion of p32/C1qbp (p32cKO) in mice caused anemia and B-lymphopenia without reduction of hematopoietic stem/progenitor cells. In addition, p32cKO mice were susceptible to hematopoietic stress with delayed recovery from anemia. p32/C1qbp-deficient CD51- TNCs exhibited impaired mitochondrial oxidation that consequently led to inactivation of mTORC1 signaling, which is essential for erythropoiesis. These findings uncover the importance of mitochondria, especially at the stage of TNCs during erythropoiesis, suggesting that dysregulation of mitochondrial protein synthesis is a cause of anemia and B-lymphopenia with an unknown pathology.

15.
Cell Rep ; 28(1): 145-158.e9, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269436

RESUMEN

Hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis by remaining quiescent in the bone marrow niche. Recapitulation of a quiescent state in culture has not been achieved, as cells rapidly proliferate and differentiate in vitro. After exhaustive analysis of different environmental factor combinations and concentrations as a way to mimic physiological conditions, we were able to maintain engraftable quiescent HSCs for 1 month in culture under very low cytokine concentrations, hypoxia, and very high fatty acid levels. Exogenous fatty acids were required likely due to suppression of intrinsic fatty acid synthesis by hypoxia and low cytokine conditions. By contrast, high cytokine concentrations or normoxia induced HSC proliferation and differentiation. Our culture system provides a means to evaluate properties of steady-state HSCs and test effects of defined factors in vitro under near-physiological conditions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Citocinas/farmacología , Ácidos Grasos/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Apoptosis , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Hipoxia de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Colesterol/farmacología , Ontología de Genes , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Humanos , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de la Célula Individual , Factor de Células Madre/farmacología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/fisiología
16.
Nat Commun ; 10(1): 1072, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842549

RESUMEN

The balance between self-renewal and differentiation of hematopoietic stem and progenitor cells (HSPCs) maintains hematopoietic homeostasis, failure of which can lead to hematopoietic disorder. HSPC fate is controlled by signals from the bone marrow niche resulting in alteration of the stem cell transcription network. Regnase-1, a member of the CCCH zinc finger protein family possessing RNAse activity, mediates post-transcriptional regulatory activity through degradation of target mRNAs. The precise function of Regnase-1 has been explored in inflammation-related cytokine expression but its function in hematopoiesis has not been elucidated. Here, we show that Regnase-1 regulates self-renewal of HSPCs through modulating the stability of Gata2 and Tal1 mRNA. In addition, we found that dysfunction of Regnase-1 leads to the rapid onset of abnormal hematopoiesis. Thus, our data reveal that Regnase-1-mediated post-transcriptional regulation is required for HSPC maintenance and suggest that it represents a leukemia tumor suppressor.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide Aguda/genética , Procesamiento Postranscripcional del ARN/fisiología , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Médula Ósea/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Autorrenovación de las Células/genética , Conjuntos de Datos como Asunto , Factor de Transcripción GATA2/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ribonucleasas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Factores de Transcripción/genética , Quimera por Trasplante
17.
Sci Adv ; 5(1): eaau9060, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30746467

RESUMEN

Compounds targeting the circadian clock have been identified as potential treatments for clock-related diseases, including cancer. Our cell-based phenotypic screen revealed uncharacterized clock-modulating compounds. Through affinity-based target deconvolution, we identified GO289, which strongly lengthened circadian period, as a potent and selective inhibitor of CK2. Phosphoproteomics identified multiple phosphorylation sites inhibited by GO289 on clock proteins, including PER2 S693. Furthermore, GO289 exhibited cell type-dependent inhibition of cancer cell growth that correlated with cellular clock function. The x-ray crystal structure of the CK2α-GO289 complex revealed critical interactions between GO289 and CK2-specific residues and no direct interaction of GO289 with the hinge region that is highly conserved among kinases. The discovery of GO289 provides a direct link between the circadian clock and cancer regulation and reveals unique design principles underlying kinase selectivity.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proliferación Celular/efectos de los fármacos , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Renales/metabolismo , Animales , Proteínas CLOCK/metabolismo , Carcinoma de Células Renales/patología , Quinasa de la Caseína II/antagonistas & inhibidores , Línea Celular Tumoral , Cristalografía por Rayos X , Células HEK293 , Humanos , Neoplasias Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación/efectos de los fármacos
18.
Int J Hematol ; 107(6): 624-626, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29728979

RESUMEN

Stem cells are characterized by their unique ability to both self-renew and differentiate along multiple cellular lineages. Self-renewal and differentiation must be tightly controlled to ensure an appropriate stem cell pool in tissue over the lifetime of an organism. Elucidating the mechanisms controlling stem cell fate and maintenance remains a key challenge in stem cell biology. Hematopoietic stem cells (HSCs) are responsible for the lifelong production of multiple blood cell lineages. To remain functional, these cells must interact with a particular microenvironment, known as the stem cell niche. HSC niches provide various factors, including cytokines, extracellular matrices, nutrients, hormones, and metabolites. These niche factors modulate cell-intrinsic molecular regulatory networks in HSCs. Niche signals also play crucial roles in the induction of HSCs from pluripotent stem cells or vascular endothelial cells. The Progress in Hematology review series in the current issue highlights some critical regulators of HSC maintenance and production.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Transducción de Señal/fisiología , Diferenciación Celular , Citocinas/fisiología , Estrés del Retículo Endoplásmico/fisiología , Matriz Extracelular/fisiología , Alimentos , Hormonas/fisiología , Humanos , Nicho de Células Madre , Respuesta de Proteína Desplegada/fisiología
19.
Cell Stem Cell ; 22(5): 713-725.e8, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29706577

RESUMEN

Stem cell self-renewal is critical for tissue homeostasis, and its dysregulation can lead to organ failure or tumorigenesis. While obesity can induce varied abnormalities in bone marrow components, it is unclear how diet might affect hematopoietic stem cell (HSC) self-renewal. Here, we show that Spred1, a negative regulator of RAS-MAPK signaling, safeguards HSC homeostasis in animals fed a high-fat diet (HFD). Under steady-state conditions, Spred1 negatively regulates HSC self-renewal and fitness, in part through Rho kinase activity. Spred1 deficiency mitigates HSC failure induced by infection mimetics and prolongs HSC lifespan, but it does not initiate leukemogenesis due to compensatory upregulation of Spred2. In contrast, HFD induces ERK hyperactivation and aberrant self-renewal in Spred1-deficient HSCs, resulting in functional HSC failure, severe anemia, and myeloproliferative neoplasm-like disease. HFD-induced hematopoietic abnormalities are mediated partly through alterations to the gut microbiota. Together, these findings reveal that diet-induced stress disrupts fine-tuning of Spred1-mediated signals to govern HSC homeostasis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Estrés Oxidativo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas Represoras/deficiencia
20.
Int J Hematol ; 107(6): 646-655, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29550946

RESUMEN

In order to maintain the homeostasis of the hematopoietic system, hematopoietic stem cells (HSCs) need to be maintained while slowly dividing over their lifetime. However, repeated cell divisions lead to the gradual accumulation of DNA damage and ultimately impair HSC function. Since telomeres are particularly fragile when subjected to replication stress, cells have several defense machinery to protect telomeres. Moreover, HSCs must protect their genome against possible DNA damage, while maintaining telomere length. A group of proteins called the shelterin complex are deeply involved in this two-way role, and it is highly resistant to the replication stress to which HSCs are subjected. Most shelterin-deficient experimental models suffer acute cytotoxicity and severe phenotypes, as each shelterin component is essential for telomere protection. The Tin2 point mutant mice show a dyskeratosis congenita (DC) like phenotype, and the Tpp1 deletion impairs the hematopoietic system. POT1/Pot1a is highly expressed in HSCs and contributes to the maintenance of the HSC pool during in vitro culture. Here, we discuss the role of shelterin molecules in HSC regulation and review current understanding of how these are regulated in the maintenance of the HSC pool and the development of hematological disorders.


Asunto(s)
Hematopoyesis/genética , Células Madre Hematopoyéticas , Proteínas de Unión a Telómeros/fisiología , Telómero , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Aminopeptidasas/genética , Animales , División Celular/genética , Daño del ADN , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Disqueratosis Congénita , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Ratones , Mutación , Serina Proteasas/genética , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...