Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 5(6): 1605-1616, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33710339

RESUMEN

Hematopoietic cell transplantation is a critical curative approach for many blood disorders. However, obtaining grafts with sufficient numbers of hematopoietic stem cells (HSCs) that maintain long-term engraftment remains challenging; this is due partly to metabolic modulations that restrict the potency of HSCs outside of their native environment. To address this, we focused on mitochondria. We found that human HSCs are heterogeneous in their mitochondrial activity as measured by mitochondrial membrane potential (MMP) even within the highly purified CD34+CD38-CD45RA-CD90+CD49f+ HSC population. We further found that the most potent HSCs exhibit the lowest mitochondrial activity in the population. We showed that the frequency of long-term culture initiating cells in MMP-low is significantly greater than in MMP-high CD34+CD38-CD45RA-CD90+ (CD90+) HSCs. Notably, these 2 populations were distinct in their long-term repopulating capacity when transplanted into immunodeficient mice. The level of chimerism 7 months posttransplantation was >50-fold higher in the blood of MMP-low relative to MMP-high CD90+ HSC recipients. Although more than 90% of both HSC subsets were in G0, MMP-low CD90+ HSCs exhibited delayed cell-cycle priming profile relative to MMP-high HSCs. These functional differences were associated with distinct mitochondrial morphology; MMP-low in contrast to MMP-high HSCs contained fragmented mitochondria. Our findings suggest that the lowest MMP level selects for the most potent, likely dormant, stem cells within the highly purified HSC population. These results identify a new approach for isolating highly potent human HSCs for further clinical applications. They also implicate mitochondria in the intrinsic regulation of human HSC quiescence and potency.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Animales , Ciclo Celular , Quimerismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Mitocondrias
2.
Stem Cell Reports ; 14(4): 561-574, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32243840

RESUMEN

Hematopoietic stem cells (HSCs) exist in a dormant state and progressively lose regenerative potency as they undergo successive divisions. Why this functional decline occurs and how this information is encoded is unclear. To better understand how this information is stored, we performed RNA sequencing on HSC populations differing only in their divisional history. Comparative analysis revealed that genes upregulated with divisions are enriched for lineage genes and regulated by cell-cycle-associated transcription factors, suggesting that proliferation itself drives lineage priming. Downregulated genes are, however, associated with an HSC signature and targeted by the Polycomb Repressive Complex 2 (PRC2). The PRC2 catalytic subunits Ezh1 and Ezh2 promote and suppress the HSC state, respectively, and successive divisions cause a switch from Ezh1 to Ezh2 dominance. We propose that cell divisions drive lineage priming and Ezh2 accumulation, which represses HSC signature genes to consolidate information on divisional history into memory.


Asunto(s)
División Celular , Linaje de la Célula , Hematopoyesis , Células Madre Hematopoyéticas/citología , Animales , División Celular/genética , Linaje de la Célula/genética , Autorrenovación de las Células , Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Regulación de la Expresión Génica , Hematopoyesis/genética , Homeostasis , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
3.
Cell Stem Cell ; 26(3): 359-376.e7, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109377

RESUMEN

Quiescence is a fundamental property that maintains hematopoietic stem cell (HSC) potency throughout life. Quiescent HSCs are thought to rely on glycolysis for their energy, but the overall metabolic properties of HSCs remain elusive. Using combined approaches, including single-cell RNA sequencing (RNA-seq), we show that mitochondrial membrane potential (MMP) distinguishes quiescent from cycling-primed HSCs. We found that primed, but not quiescent, HSCs relied readily on glycolysis. Notably, in vivo inhibition of glycolysis enhanced the competitive repopulation ability of primed HSCs. We further show that HSC quiescence is maintained by an abundance of large lysosomes. Repression of lysosomal activation in HSCs led to further enlargement of lysosomes while suppressing glucose uptake. This also induced increased lysosomal sequestration of mitochondria and enhanced the competitive repopulation ability of primed HSCs by over 90-fold in vivo. These findings show that restraining lysosomal activity preserves HSC quiescence and potency and may be therapeutically relevant.


Asunto(s)
Células Madre Hematopoyéticas , Mitocondrias , División Celular , Glucólisis , Células Madre Hematopoyéticas/metabolismo , Lisosomas , Mitocondrias/metabolismo
4.
Ann N Y Acad Sci ; 1466(1): 59-72, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31621095

RESUMEN

The induction of hematopoiesis in various cell types via transcription factor (TF) reprogramming has been demonstrated by several strategies. The eventual goal of these approaches is to generate a product for unmet needs in hematopoietic cell transplantation therapies. The most successful strategies hew closely to clues provided from developmental hematopoiesis in terms of factor expression and environmental cues. In this review, we aim to summarize the TFs that play important roles in developmental hematopoiesis primarily and to also touch on adult hematopoiesis. Several aspects of cellular and molecular biology coalesce in this process, with TFs and surrounding cellular signals playing a major role in the overall development of the hematopoietic lineage. We attempt to put these elements into the context of reprogramming and highlight their roles.


Asunto(s)
Microambiente Celular/fisiología , Crecimiento y Desarrollo/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción/fisiología , Animales , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Crecimiento y Desarrollo/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Humanos , Nicho de Células Madre/fisiología
5.
J Vis Exp ; (153)2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31736500

RESUMEN

The cellular and molecular mechanisms underlying specification of human hematopoietic stem cells (HSCs) remain elusive. Strategies to recapitulate human HSC emergence in vitro are required to overcome limitations in studying this complex developmental process. Here, we describe a protocol to generate hematopoietic stem and progenitor-like cells from human dermal fibroblasts employing a direct cell reprogramming approach. These cells transit through a hemogenic intermediate cell-type, resembling the endothelial-to-hematopoietic transition (EHT) characteristic of HSC specification. Fibroblasts were reprogrammed to hemogenic cells via transduction with GATA2, GFI1B and FOS transcription factors. This combination of three factors induced morphological changes, expression of hemogenic and hematopoietic markers and dynamic EHT transcriptional programs. Reprogrammed cells generate hematopoietic progeny and repopulate immunodeficient mice for three months. This protocol can be adapted towards the mechanistic dissection of the human EHT process as exemplified here by defining GATA2 targets during the early phases of reprogramming. Thus, human hemogenic reprogramming provides a simple and tractable approach to identify novel markers and regulators of human HSC emergence. In the future, faithful induction of hemogenic fate in fibroblasts may lead to the generation of patient-specific HSCs for transplantation.


Asunto(s)
Reprogramación Celular , Fibroblastos/fisiología , Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción/metabolismo , Animales , Adhesión Celular , Diferenciación Celular , Humanos , Ratones , Factores de Transcripción/genética
6.
FEBS Lett ; 593(23): 3266-3287, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31557312

RESUMEN

Transcription factor (TF)-based reprogramming of somatic tissues holds great promise for regenerative medicine. Previously, we demonstrated that the TFs GATA2, GFI1B, and FOS convert mouse and human fibroblasts to hemogenic endothelial-like precursors that generate hematopoietic stem progenitor (HSPC)-like cells over time. This conversion is lacking in robustness both in yield and biological function. Herein, we show that inclusion of GFI1 to the reprogramming cocktail significantly expands the HSPC-like population. AFT024 coculture imparts functional potential to these cells and allows quantification of stem cell frequency. Altogether, we demonstrate an improved human hemogenic induction protocol that could provide a valuable human in vitro model of hematopoiesis for disease modeling and a platform for cell-based therapeutics. DATABASE: Gene expression data are available in the Gene Expression Omnibus (GEO) database under the accession number GSE130361.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Animales , Técnicas de Cocultivo/métodos , Fibroblastos/citología , Fibroblastos/metabolismo , Factor de Transcripción GATA2/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Represoras/genética , Factores de Transcripción/genética
7.
Cell Rep ; 28(6): 1400-1409.e4, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390555

RESUMEN

A multitude of signals are coordinated to maintain self-renewal in embryonic stem cells (ESCs). To unravel the essential internal and external signals required for sustaining the ESC state, we expand upon a set of ESC pluripotency-associated phosphoregulators (PRs) identified previously by short hairpin RNA (shRNA) screening. In addition to the previously described Aurka, we identify 4 additional PRs (Bub1b, Chek1, Ppm1g, and Ppp2r1b) whose depletion compromises self-renewal and leads to consequent differentiation. Global gene expression profiling and computational analyses reveal that knockdown of the 5 PRs leads to DNA damage/genome instability, activating p53 and culminating in ESC differentiation. Similarly, depletion of genome integrity-associated genes involved in DNA replication and checkpoint, mRNA processing, and Charcot-Marie-Tooth disease lead to compromise of ESC self-renewal via an increase in p53 activity. Our studies demonstrate an essential link between genomic integrity and developmental cell fate regulation in ESCs.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/fisiología , Inestabilidad Genómica , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Línea Celular , Daño del ADN , Perfilación de la Expresión Génica , Prueba de Complementación Genética , Ratones , Fosfoproteínas/genética , Fosfoproteínas/fisiología , ARN Interferente Pequeño , Transducción de Señal , Proteína p53 Supresora de Tumor/fisiología
8.
Cell Rep ; 25(10): 2821-2835.e7, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517869

RESUMEN

During development, hematopoietic stem and progenitor cells (HSPCs) arise from specialized endothelial cells by a process termed endothelial-to-hematopoietic transition (EHT). The genetic program driving human HSPC emergence remains largely unknown. We previously reported that the generation of hemogenic precursor cells from mouse fibroblasts recapitulates developmental hematopoiesis. Here, we demonstrate that human fibroblasts can be reprogrammed into hemogenic cells by the same transcription factors. Induced cells display dynamic EHT transcriptional programs, generate hematopoietic progeny, possess HSPC cell surface phenotype, and repopulate immunodeficient mice for 3 months. Mechanistically, GATA2 and GFI1B interact and co-occupy a cohort of targets. This cooperative binding is reflected by engagement of open enhancers and promoters, initiating silencing of fibroblast genes and activating the hemogenic program. However, GATA2 displays dominant and independent targeting activity during the early phases of reprogramming. These findings shed light on the processes controlling human HSC specification and support generation of reprogrammed HSCs for clinical applications.


Asunto(s)
Reprogramación Celular , Hemangioblastos/citología , Hemangioblastos/metabolismo , Factores de Transcripción/metabolismo , Adulto , Secuencia de Bases , Elementos de Facilitación Genéticos/genética , Fibroblastos/metabolismo , Factor de Transcripción GATA2/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Recién Nacido , Fenotipo , Regiones Promotoras Genéticas/genética , Unión Proteica
9.
Proc Natl Acad Sci U S A ; 115(47): E11128-E11137, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30385632

RESUMEN

Osteosarcoma (OS), the most common primary bone tumor, is highly metastatic with high chemotherapeutic resistance and poor survival rates. Using induced pluripotent stem cells (iPSCs) generated from Li-Fraumeni syndrome (LFS) patients, we investigate an oncogenic role of secreted frizzled-related protein 2 (SFRP2) in p53 mutation-associated OS development. Interestingly, we find that high SFRP2 expression in OS patient samples correlates with poor survival. Systems-level analyses identified that expression of SFRP2 increases during LFS OS development and can induce angiogenesis. Ectopic SFRP2 overexpression in normal osteoblast precursors is sufficient to suppress normal osteoblast differentiation and to promote OS phenotypes through induction of oncogenic molecules such as FOXM1 and CYR61 in a ß-catenin-independent manner. Conversely, inhibition of SFRP2, FOXM1, or CYR61 represses the tumorigenic potential. In summary, these findings demonstrate the oncogenic role of SFRP2 in the development of p53 mutation-associated OS and that inhibition of SFRP2 is a potential therapeutic strategy.


Asunto(s)
Neoplasias Óseas/genética , Carcinogénesis/genética , Síndrome de Li-Fraumeni/patología , Proteínas de la Membrana/genética , Osteosarcoma/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Proteína 61 Rica en Cisteína/antagonistas & inhibidores , Proteína 61 Rica en Cisteína/genética , Proteína Forkhead Box M1/antagonistas & inhibidores , Proteína Forkhead Box M1/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de Li-Fraumeni/genética , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Osteoblastos/citología , Osteosarcoma/patología
10.
PLoS One ; 13(10): e0203597, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30289930

RESUMEN

Research in photobiology is currently limited by a lack of devices capable of delivering precise and tunable irradiation to cells in a high-throughput format. This limits researchers to using expensive commercially available or custom-built light sources which make it difficult to replicate, standardize, optimize, and scale experiments. Here we present an open-source Microplate Photoirradiation System (MPS) developed to enable high-throughput light experiments in standard 96 and 24-well microplates for a variety of applications in photobiology research. This open-source system features 96 independently controlled LEDs (4 LEDs per well in 24-well), Wi-Fi connected control and programmable graphical user interface (GUI) for control and programming, automated calibration GUI, and modular control and LED boards for maximum flexibility. A web-based GUI generates light program files containing irradiation parameters for groups of LEDs. These parameters are then uploaded wirelessly, stored and used on the MPS to run photoirradiation experiments inside any incubator. A rapid and semi-quantitative porphyrin metabolism assay was also developed to validate the system in wild-type fibroblasts. Protoporphyrin IX (PpIX) fluorescence accumulation was induced by incubation with 5-aminolevulinic acid (ALA), a photosensitization method leveraged clinically to destroy malignant cell types in a process termed photodynamic therapy (PDT), and cells were irradiated with 405nm light with varying irradiance, duration and pulsation parameters. Immediately after light treatment with the MPS, subsequent photobleaching was measured in live, adherent cells in both 96-well and a 24-well microplates using a microplate reader. Results demonstrate the utility and reliability of the Microplate Photoirradiation System to irradiate cells with precise irradiance and timing parameters in order to measure PpIx photobleaching kinetics in live adherent cells and perform comparable experiments with both 24 and 96 well microplate formats. The high-throughput capability of the MPS enabled measurement of enough irradiance conditions in a single microplate to fit PpIX fluorescence to a bioexponential decay model of photobleaching, as well as reveal a dependency of photobleaching on duty-cycle-but not frequency-in a pulsed irradiance regimen.


Asunto(s)
Fotobiología/métodos , Fotoquimioterapia/métodos , Trastornos por Fotosensibilidad , Protoporfirinas/química , Ácido Aminolevulínico/química , Ácido Aminolevulínico/efectos de la radiación , Gráficos por Computador , Humanos , Luz , Fotoblanqueo , Protoporfirinas/efectos de la radiación , Radiación , Tecnología Inalámbrica
11.
Blood ; 129(14): 1901-1912, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28179275

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is used clinically to treat leukopenia and to enforce hematopoietic stem cell (HSC) mobilization to the peripheral blood (PB). However, G-CSF is also produced in response to infection, and excessive exposure reduces HSC repopulation capacity. Previous work has shown that dormant HSCs contain all the long-term repopulation potential in the bone marrow (BM), and that as HSCs accumulate a divisional history, they progressively lose regenerative potential. As G-CSF treatment also induces HSC proliferation, we sought to examine whether G-CSF-mediated repopulation defects are a result of increased proliferative history. To do so, we used an established H2BGFP label retaining system to track HSC divisions in response to G-CSF. Our results show that dormant HSCs are preferentially mobilized to the PB on G-CSF treatment. We find that this mobilization does not result in H2BGFP label dilution of dormant HSCs, suggesting that G-CSF does not stimulate dormant HSC proliferation. Instead, we find that proliferation within the HSC compartment is restricted to CD41-expressing cells that function with short-term, and primarily myeloid, regenerative potential. Finally, we show CD41 expression is up-regulated within the BM HSC compartment in response to G-CSF treatment. This emergent CD41Hi HSC fraction demonstrates no observable engraftment potential, but directly matures into megakaryocytes when placed in culture. Together, our results demonstrate that dormant HSCs mobilize in response to G-CSF treatment without dividing, and that G-CSF-mediated proliferation is restricted to cells with limited regenerative potential found within the HSC compartment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/biosíntesis , Animales , Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Ratones , Ratones Transgénicos , Glicoproteína IIb de Membrana Plaquetaria/genética
12.
Cell ; 167(5): 1296-1309.e10, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27839867

RESUMEN

The ability of cells to count and remember their divisions could underlie many alterations that occur during development, aging, and disease. We tracked the cumulative divisional history of slow-cycling hematopoietic stem cells (HSCs) throughout adult life. This revealed a fraction of rarely dividing HSCs that contained all the long-term HSC (LT-HSC) activity within the aging HSC compartment. During adult life, this population asynchronously completes four traceable symmetric self-renewal divisions to expand its size before entering a state of dormancy. We show that the mechanism of expansion involves progressively lengthening periods between cell divisions, with long-term regenerative potential lost upon a fifth division. Our data also show that age-related phenotypic changes within the HSC compartment are divisional history dependent. These results suggest that HSCs accumulate discrete memory stages over their divisional history and provide evidence for the role of cellular memory in HSC aging.


Asunto(s)
Envejecimiento/patología , Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Animales , Trasplante de Médula Ósea , Ciclo Celular , División Celular , Ratones , Ratones Endogámicos C57BL , Glicoproteína IIb de Membrana Plaquetaria/metabolismo
13.
Blood ; 128(9): 1181-92, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27365423

RESUMEN

The maintenance of hematopoietic stem cells (HSCs) during ex vivo culture is an important prerequisite for their therapeutic manipulation. However, despite intense research, culture conditions for robust maintenance of HSCs are still missing. Cultured HSCs are quickly lost, preventing their improved analysis and manipulation. Identification of novel factors supporting HSC ex vivo maintenance is therefore necessary. Coculture with the AFT024 stroma cell line is capable of maintaining HSCs ex vivo long-term, but the responsible molecular players remain unknown. Here, we use continuous long-term single-cell observation to identify the HSC behavioral signature under supportive or nonsupportive stroma cocultures. We report early HSC survival as a major characteristic of HSC-maintaining conditions. Behavioral screening after manipulation of candidate molecules revealed that the extracellular matrix protein dermatopontin (Dpt) is involved in HSC maintenance. DPT knockdown in supportive stroma impaired HSC survival, whereas ectopic expression of the Dpt gene or protein in nonsupportive conditions restored HSC survival. Supplementing defined stroma- and serum-free culture conditions with recombinant DPT protein improved HSC clonogenicity. These findings illustrate a previously uncharacterized role of Dpt in maintaining HSCs ex vivo.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Células Madre Hematopoyéticas/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/farmacología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/farmacología , Células Madre Hematopoyéticas/citología , Masculino , Ratones , Ratones Transgénicos , Células del Estroma/citología , Células del Estroma/metabolismo , Factores de Tiempo
14.
Dev Cell ; 36(5): 525-39, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26954547

RESUMEN

Definitive hematopoiesis emerges via an endothelial-to-hematopoietic transition in the embryo and placenta; however, the precursor cells to hemogenic endothelium are not defined phenotypically. We previously demonstrated that the induction of hematopoietic progenitors from fibroblasts progresses through hemogenic precursors that are Prom1(+)Sca1(+)CD34(+)CD45(-) (PS34CD45(-)). Guided by these studies, we analyzed mouse placentas and identified a population with this phenotype. These cells express endothelial markers, are heterogeneous for early hematopoietic markers, and localize to the vascular labyrinth. Remarkably, global gene expression profiles of PS34CD45(-) cells correlate with reprogrammed precursors and establish a hemogenic precursor cell molecular signature. PS34CD45(-) cells are also present in intra-embryonic hemogenic sites. After stromal co-culture, PS34CD45(-) cells give rise to all blood lineages and engraft primary and secondary immunodeficient mice. In summary, we show that reprogramming reveals a phenotype for in vivo precursors to hemogenic endothelium, establishing that direct in vitro conversion informs developmental processes in vivo.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Reprogramación Celular , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Embrionarias de Ratones/citología , Animales , Células Cultivadas , Endotelio/metabolismo , Femenino , Fibroblastos/citología , Ratones , Ratones Endogámicos C57BL , Embarazo
15.
Ann N Y Acad Sci ; 1370(1): 24-35, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26748878

RESUMEN

Even though all paradigms of stem cell therapy and regenerative medicine emerged from the study of hematopoietic stem cells (HSCs), the inability to generate these cells de novo or expand them in vitro persists. Initial efforts to obtain these cells began with the use of embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) technologies, but these strategies have yet to yield fully functional cells. Subsequently, more recent approaches involve transcription factor (TF) overexpression to reprogram PSCs and various somatic cells. The induction of pluripotency with just four TFs by Yamanaka informs our ability to convert cell fates and demonstrates the feasibility of utilizing terminally differentiated cells to generate cells with multilineage potential. In this review, we discuss the recent efforts undertaken using TF-based reprogramming strategies to convert several cell types into HSCs.


Asunto(s)
Linaje de la Célula/fisiología , Células Madre Hematopoyéticas/metabolismo , Trasplante de Células Madre/métodos , Factores de Transcripción/metabolismo , Animales , Células Madre Hematopoyéticas/citología , Humanos , Medicina Regenerativa
16.
Trends Cell Biol ; 26(3): 202-214, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26526106

RESUMEN

Previous attempts to either generate or expand hematopoietic stem cells (HSCs) in vitro have involved either ex vivo expansion of pre-existing patient or donor HSCs or de novo generation from pluripotent stem cells (PSCs), comprising both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). iPSCs alleviated ESC ethical issues but attempts to generate functional mature hematopoietic stem and progenitor cells (HSPCs) have been largely unsuccessful. New efforts focus on directly reprogramming somatic cells into definitive HSCs and HSPCs. To meet clinical needs and to advance drug discovery and stem cell therapy, alternative approaches are necessary. In this review, we synthesize the strategies used and the key findings made in recent years by those trying to make an HSC.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Reprogramación Celular , Células Madre Embrionarias/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología
17.
J Vis Exp ; (118)2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-28060340

RESUMEN

This protocol details the induction of a hemogenic program in mouse embryonic fibroblasts (MEFs) via overexpression of transcription factors (TFs). We first designed a reporter screen using MEFs from human CD34-tTA/TetO-H2BGFP (34/H2BGFP) double transgenic mice. CD34+ cells from these mice label H2B histones with GFP, and cease labeling upon addition of doxycycline (DOX). MEFS were transduced with candidate TFs and then observed for the emergence of GFP+ cells that would indicate the acquisition of a hematopoietic or endothelial cell fate. Starting with 18 candidate TFs, and through a process of combinatorial elimination, we obtained a minimal set of factors that would induce the highest percentage of GFP+ cells. We found that Gata2, Gfi1b, and cFos were necessary and the addition of Etv6 provided the optimal induction. A series of gene expression analyses done at different time points during the reprogramming process revealed that these cells appeared to go through a precursor cell that underwent an endothelial to hematopoietic transition (EHT). As such, this reprogramming process mimics developmental hematopoiesis "in a dish," allowing study of hematopoiesis in vitro and a platform to identify the mechanisms that underlie this specification. This methodology also provides a framework for translation of this work to the human system in the hopes of generating an alternative source of patient-specific hematopoietic stem cells (HSCs) for a number of applications in the treatment and study of hematologic diseases.


Asunto(s)
Fibroblastos/fisiología , Hematopoyesis/genética , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Fibroblastos/citología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas , Ratones
18.
Cell Rep ; 13(3): 504-515, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26456833

RESUMEN

Somatic PTPN11 mutations cause juvenile myelomonocytic leukemia (JMML). Germline PTPN11 defects cause Noonan syndrome (NS), and specific inherited mutations cause NS/JMML. Here, we report that hematopoietic cells differentiated from human induced pluripotent stem cells (hiPSCs) harboring NS/JMML-causing PTPN11 mutations recapitulated JMML features. hiPSC-derived NS/JMML myeloid cells exhibited increased signaling through STAT5 and upregulation of miR-223 and miR-15a. Similarly, miR-223 and miR-15a were upregulated in 11/19 JMML bone marrow mononuclear cells harboring PTPN11 mutations, but not those without PTPN11 defects. Reducing miR-223's function in NS/JMML hiPSCs normalized myelogenesis. MicroRNA target gene expression levels were reduced in hiPSC-derived myeloid cells as well as in JMML cells with PTPN11 mutations. Thus, studying an inherited human cancer syndrome with hiPSCs illuminated early oncogenesis prior to the accumulation of secondary genomic alterations, enabling us to discover microRNA dysregulation, establishing a genotype-phenotype association for JMML and providing therapeutic targets.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Leucemia Mielomonocítica Juvenil/metabolismo , Células Mieloides/citología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patología , MicroARNs/genética , Mutación , Células Mieloides/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Regulación hacia Arriba
19.
BMC Cancer ; 15: 444, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26025442

RESUMEN

BACKGROUND: The mechanisms allowing residual multiple myeloma (MM) cells to persist after bortezomib (Bz) treatment remain unclear. We hypothesized that studying the biology of bortezomib-surviving cells may reveal markers to identify these cells and survival signals to target and kill residual MM cells. METHODS: We used H2B-GFP label retention, biochemical tools and in vitro and in vivo experiments to characterize growth arrest and the unfolded protein responses in quiescent Bz-surviving cells. We also tested the effect of a demethylating agent, 5-Azacytidine, on Bz-induced quiescence and whether inhibiting the chaperone GRP78/BiP (henceforth GRP78) with a specific toxin induced apoptosis in Bz-surviving cells. Finally, we used MM patient samples to test whether GRP78 levels might associate with disease progression. Statistical analysis employed t-test and Mann-Whitney tests at a 95% confidence. RESULTS: We report that Bz-surviving MM cells in vitro and in vivo enter quiescence characterized by p21(CIP1) upregulation. Bz-surviving MM cells also downregulated CDK6, Ki67 and P-Rb. H2B-GFP label retention showed that Bz-surviving MM cells are either slow-cycling or deeply quiescent. The Bz-induced quiescence was stabilized by low dose (500nM) of 5-azacytidine (Aza) pre-treatment, which also potentiated the initial Bz-induced apoptosis. We also found that expression of GRP78, an unfolded protein response (UPR) survival factor, persisted in MM quiescent cells. Importantly, GRP78 downregulation using a specific SubAB bacterial toxin killed Bz-surviving MM cells. Finally, quantification of Grp78(high)/CD138+ MM cells from patients suggested that high levels correlated with progressive disease. CONCLUSIONS: We conclude that Bz-surviving MM cells display a GRP78(HIGH)/p21(HIGH)/CDK6(LOW)/P-Rb(LOW) profile, and these markers may identify quiescent MM cells capable of fueling recurrences. We further conclude that Aza + Bz treatment of MM may represent a novel strategy to delay recurrences by enhancing Bz-induced apoptosis and quiescence stability.


Asunto(s)
Bortezomib/administración & dosificación , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Proteínas de Choque Térmico/biosíntesis , Mieloma Múltiple/tratamiento farmacológico , Quinasas p21 Activadas/biosíntesis , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Azacitidina/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/genética , Chaperón BiP del Retículo Endoplásmico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/genética
20.
NPJ Syst Biol Appl ; 1: 15012, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-28725462

RESUMEN

BACKGROUND: Stem cell differentiation is a complex biological process. Cellular heterogeneity, such as the co-existence of different cell subpopulations within a population, partly hampers our understanding of this process. The modern single-cell gene expression technologies, such as single-cell RT-PCR and RNA-seq, have enabled us to elucidate such heterogeneous cell subpopulations. However, the identification of a transcriptional regulatory network (TRN) for each cell subpopulation within a population and genes determining specific cell fates (lineage specifiers) remains a challenge due to the slower development of appropriate computational and experimental workflows. Here, we propose a computational differential network analysis approach for predicting lineage specifiers in binary-fate differentiation events. METHODS: The proposed method is based on a model that considers each stem cell subpopulation being in a stable state maintained by its specific TRN stability core, and cell differentiation involves changes in these stability cores between parental and daughter cell subpopulations. The method first reconstructs topologically different cell-subpopulation specific TRNs from single-cell gene expression data, literature knowledge and transcription factor (TF)-DNA binding-site prediction. Then, it systematically predicts lineage specifiers by identifying genes in the TRN stability cores in both parental and daughter cell subpopulations. RESULTS: Application of this method to different stem cell differentiation systems was able to predict known and putative novel lineage specifiers. These examples include the differentiation of inner cell mass into either primitive endoderm or epiblast, different progenitor cells in the hematopoietic system, and the lung alveolar bipotential progenitor into either alveolar type 1 or alveolar type 2. CONCLUSIONS: The method is generally applicable to any binary-fate differentiation system, for which single-cell gene expression data are available. Therefore, it should aid in understanding stem cell lineage specification, and in the development of experimental strategies for regenerative medicine.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA