Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(5): 690-703, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34657154

RESUMEN

The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.


Asunto(s)
Autofagia , Trastornos de Fallo de la Médula Ósea/metabolismo , Hematopoyesis , Células Madre Mesenquimatosas/citología , Animales , Células Cultivadas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Estrés Oxidativo , Proteína Wnt-5a/metabolismo
2.
Haematologica ; 108(2): 490-501, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35950533

RESUMEN

Remodeling of the bone marrow microenvironment in chronic inflammation and in aging reduces hematopoietic stem cell (HSC) function. To assess the mechanisms of this functional decline of HSC and find strategies to counteract it, we established a model in which the Sfrp1 gene was deleted in Osterix+ osteolineage cells (OS1Δ/Δ mice). HSC from these mice showed severely diminished repopulating activity with associated DNA damage, enriched expression of the reactive oxygen species pathway and reduced single-cell proliferation. Interestingly, not only was the protein level of Catenin beta-1 (bcatenin) elevated, but so was its association with the phosphorylated co-activator p300 in the nucleus. Since these two proteins play a key role in promotion of differentiation and senescence, we inhibited in vivo phosphorylation of p300 through PP2A-PR72/130 by administration of IQ-1 in OS1Δ/Δ mice. This treatment not only reduced the b-catenin/phosphop300 association, but also decreased nuclear p300. More importantly, in vivo IQ-1 treatment fully restored HSC repopulating activity of the OS1Δ/Δ mice. Our findings show that the osteoprogenitor Sfrp1 is essential for maintaining HSC function. Furthermore, pharmacological downregulation of the nuclear b-catenin/phospho-p300 association is a new strategy to restore poor HSC function.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Ratones , Animales , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Médula Ósea/metabolismo , Envejecimiento , Especies Reactivas de Oxígeno/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
3.
Stem Cells ; 39(6): 819-830, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33539629

RESUMEN

Survival of chronic lymphocytic leukemia (CLL) cells critically depends on the support of an adapted and therefore appropriate tumor microenvironment. Increasing evidence suggests that B-cell receptor-associated kinases such as protein kinase C-ß (PKCß) or Lyn kinase are essential for the formation of a microenvironment supporting leukemic growth. Here, we describe the impact of PKCß on the glucose metabolism in bone marrow stromal cells (BMSC) upon CLL contact. BMSC get activated by CLL contact expressing stromal PKCß that diminishes mitochondrial stress and apoptosis in CLL cells by stimulating glucose uptake. In BMSC, the upregulation of PKCß results in increased mitochondrial depolarization and leads to a metabolic switch toward oxidative phosphorylation. In addition, PKCß-deficient BMSC regulates the expression of Hnf1 promoting stromal insulin signaling after CLL contact. Our data suggest that targeting PKCß and the glucose metabolism of the leukemic niche could be a potential therapeutic strategy to overcome stroma-mediated drug resistance.


Asunto(s)
Células de la Médula Ósea/metabolismo , Glucosa/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteína Quinasa C beta/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Proteína Quinasa C beta/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Microambiente Tumoral/efectos de los fármacos
4.
Blood ; 134(15): 1214-1226, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31366622

RESUMEN

A major limitation preventing in vivo modulation of hematopoietic stem cells (HSCs) is the incomplete understanding of the cellular and molecular support of the microenvironment in regulating HSC fate decisions. Consequently, murine HSCs cannot be generated, maintained, or expanded in culture over extended periods of time. A significantly improved understanding of the bone marrow niche environment and its molecular interactions with HSCs is pivotal to overcoming this challenge. We here prospectively isolated all major nonhematopoietic cellular niche components and cross-correlate them in detail with niche cells defined by lineage marking or tracing. Compiling an extensive database of soluble and membrane-bound ligand-receptor interactions, we developed a computational method to infer potential cell-to-cell interactions based on transcriptome data of sorter-purified niche cells and hematopoietic stem and progenitor cell subpopulations. Thus, we establish a compendium of the molecular communication between defined niche components and HSCs. Our analysis suggests an important role for cytokine antagonists in the regulation of HSC functions.


Asunto(s)
Células de la Médula Ósea/citología , Comunicación Celular , Células Madre Hematopoyéticas/citología , Nicho de Células Madre , Animales , Diferenciación Celular , Separación Celular , Ratones Endogámicos C57BL
5.
Haematologica ; 106(10): 2633-2640, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33543864

RESUMEN

Hematopoietic stem cell self-renewal, proliferation, and differentiation are independently regulated by intrinsic as well as extrinsic mechanisms. We previously demonstrated that murine proliferation of hematopoietic stem cells is supported in serum-free medium supplemented with two growth factors, stem cell factor and interleukin 11. The survival of hematopoietic stem cells is additionally improved by supplementing this medium with two more growth factors, neural growth factor and collagen 1 (four growth factors) or serum-free medium conditioned by the hematopoietic stem cell-supportive stromal UG26-1B6 cells1. Here, we describe a robust and versatile alternative source of conditioned medium from mouse embryonic fibroblasts. We found that this conditioned medium supports survival and phenotypical identity of hematopoietic stem cells, as well as cell cycle entry in single cell cultures of CD34- CD48- CD150+ Lineage- SCA1+ KIT+ cells supplemented with two growth factors. Strikingly, in comparison with cultures in serum-free medium with four growth factors, conditioned medium from mouse embryonic fibroblasts increases the numbers of proliferating clones and the number of Lineage- SCA1+ KIT+ cells, both with two and four growth factors. In addition, conditioned medium from mouse embryonic fibroblasts supports self-renewal in culture of cells with short- and long-term hematopoiesis-repopulating ability in vivo. These findings identify conditioned medium from mouse embryonic fibroblasts as a robust alternative serumfree source of factors to maintain self-renewal of in vivo-repopulating hematopoetic stem cells in culture.


Asunto(s)
Fibroblastos , Células Madre Hematopoyéticas , Animales , Diferenciación Celular , División Celular , Células Cultivadas , Hematopoyesis , Ratones
6.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769079

RESUMEN

The IL-6 family cytokine Oncostatin M (OSM) is involved in cell development, growth, hematopoiesis, inflammation, and cancer. Intriguingly, OSM has proliferative and antiproliferative effects depending on the target cell. The molecular mechanisms underlying these opposing effects are not fully understood. Previously, we found OSM upregulation in different myeloproliferative syndromes. However, OSM receptor (OSMR) expression was detected on stromal cells but not the malignant cells themselves. In the present study, we, therefore, investigated the effect of murine OSM (mOSM) on proliferation in stromal and fibroblast cell lines. We found that mOSM impairs the proliferation of bone marrow (BM) stromal cells, whereas fibroblasts responded to mOSM with increased proliferation. When we set out to reveal the mechanisms underlying these opposing effects, we detected increased expression of the OSM receptors OSMR and LIFR in stromal cells. Interestingly, Osmr knockdown and Lifr overexpression attenuated the OSM-mediated effect on proliferation in both cell lines indicating that mOSM affected the proliferation signaling mainly through the OSMR. Furthermore, mOSM induced activation of the JAK-STAT, PI3K-AKT, and MAPK-ERK pathways in OP9 and NIH/3T3 cells with differences in total protein levels between the two cell lines. Our findings offer new insights into the regulation of proliferation by mOSM.


Asunto(s)
Proliferación Celular , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Animales , Línea Celular , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células 3T3 NIH , Transducción de Señal
7.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24141946

RESUMEN

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Asunto(s)
Senescencia Celular , Células Madre Hematopoyéticas/citología , Vía de Señalización Wnt , Animales , Diferenciación Celular , Polaridad Celular , Femenino , Haploinsuficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Rejuvenecimiento , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Proteína de Unión al GTP cdc42/metabolismo
8.
Blood ; 126(9): 1051-2, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26316612

RESUMEN

In this issue of Blood, Kabiri and coworkers report the hematopoietic deletion of the endoplasmic reticulum­localized O-acyltransferase porcupine (PORCN), which is necessary for acylation of Wnts in the endoplasmic reticulum, enabling their secretion and binding to the frizzled receptors. Unexpectedly, the absence of secreted Wnt factors does not have major effects on steady-state in vivo hematopoiesis or on long-term repopulating activity of Wnt-deficient hematopoietic stem cells.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Vía de Señalización Wnt , Animales
9.
J Vasc Res ; 54(6): 321-328, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28950284

RESUMEN

Insulin-like growth factors (IGFs) are mediators of growth hormone-induced metabolic and anabolic actions, but also regulate cell growth, differentiation, and apoptosis and show beneficial effects in acute myocardial ischemia. Since endothelial progenitor cells (EPCs) improve myocardial function after acute myocardial infarction, we sought to investigate whether overexpression of IGF-2 in expanded EPCs (eEPCs) further contributes to improvement in left ventricular function after myocardial infarction. EPCs were isolated from human cord blood and transduced by a retroviral vector expression of IGF-2 (IGF-2 eEPCs) or vector only. Expression levels were confirmed by RT-PCR. Vector only-transduced eEPCs or IGF-2 eEPCs were transplanted after ligation of the left anterior descending coronary artery in athymic nude rats. Transplantation of eEPCs improved the left ventricular ejection fraction after 2 weeks. Overexpression of IGF-2 further improved the left ventricular ejection fraction and reduced the myocardial infarction size. Immunohistological analysis revealed an increase in proliferating cells and a decrease in monocytes and apoptotic myocytes within the infarction zone after transplantation of IGF-2-overexpressing eEPCs. Transplantation of IGF-2-overexpressing eEPCs in acute myocardial infarction may improve early myocardial function by enhancing proliferation and limiting the inflammatory response and apoptosis.


Asunto(s)
Proliferación Celular , Células Progenitoras Endoteliales/trasplante , Factor II del Crecimiento Similar a la Insulina/metabolismo , Infarto del Miocardio/cirugía , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Función Ventricular Izquierda , Animales , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Monocitos/metabolismo , Monocitos/patología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Ratas Desnudas , Recuperación de la Función , Volumen Sistólico , Factores de Tiempo , Transfección , Regulación hacia Arriba
10.
Stem Cells ; 34(9): 2381-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27299503

RESUMEN

Sfrp2 is overexpressed in stromal cells which maintain hematopoietic stem cells (HSCs) during in vitro culture. We here showed, that coculture of hematopoetic cells with stromal cells with reduced expression of Sfrp2 increases the number lineage-negative Kit(+) Sca-1(+) (LSK) and progenitor cells in vitro. The LSK cells from these cocultures showed activation of canonical Wnt signaling, higher levels of Ki-67, BrdU incorporation, and the number of γH2A.X positive foci. Total repopulating activity of these cultures was, however, diminished, indicating loss of HSC. To extend these in vitro data, we modelled stress in vivo, i.e., by aging, or 5-FU treatment in Sfrp2(-) (/) (-) mice, or replicative stress in regeneration of HSCs in Sfrp2(-) (/) (-) recipients. In all three in vivo stress situations, we noted an increase of LSK cells, characterized by increased levels of ß-catenin and cyclin D1. In the transplantation experiments, the increase in LSK cells in primary recipients was subsequently associated with a progressive loss of HSCs in serial transplantations. Similar to the in vitro coculture stress, in vivo genotoxic stress in 5-FU-treated Sfrp2(-) (/) (-) mice increased cell cycle activity of LSK cells with higher levels of BrdU incorporation, increased expression of Ki-67, and canonical Wnt signaling. Importantly, as noted in vitro, increased cycling of LSKs in vivo was accompanied by a defective γH2A.X-dependent DNA damage response and depolarized localization of acetylated H4K16. Our experiments support the view that Sfrp2 expression in the niche is required to maintain the HSC pool by limiting stress-induced DNA damage and attenuating canonical Wnt-mediated HSC activation. Stem Cells 2016;34:2381-2392.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de la Membrana/deficiencia , Regeneración , Nicho de Células Madre , Estrés Fisiológico , Envejecimiento/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Daño del ADN , Fluorouracilo/farmacología , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Regeneración/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
11.
Blood ; 124(26): 3905-13, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25352127

RESUMEN

Thymocyte selection-associated high mobility group box protein family member 2 (TOX2) is a transcription factor belonging to the TOX family that shares a highly conserved high mobility group DNA-binding domain with the other TOX members. Although TOX1 has been shown to be an essential regulator of T-cell and natural killer (NK) cell differentiation in mice, little is known about the roles of the other TOX family members in lymphocyte development, particularly in humans. In this study, we found that TOX2 was preferentially expressed in mature human NK cells (mNK) and was upregulated during in vitro differentiation of NK cells from human umbilical cord blood (UCB)-derived CD34(+) cells. Gene silencing of TOX2 intrinsically hindered the transition between early developmental stages of NK cells, whereas overexpression of TOX2 enhanced the development of mNK cells from UCB CD34(+) cells. We subsequently found that TOX2 was independent of ETS-1 but could directly upregulate the transcription of TBX21 (encoding T-BET). Overexpression of T-BET rescued the TOX2 knockdown phenotypes. Given the essential function of T-BET in NK cell differentiation, TOX2 therefore plays a crucial role in controlling normal NK cell development by acting upstream of TBX21 transcriptional regulation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/metabolismo , Células Asesinas Naturales/citología , Proteínas de Dominio T Box/metabolismo , Animales , Antígenos CD34/metabolismo , Diferenciación Celular , Sangre Fetal/citología , Silenciador del Gen , Células HEK293 , Humanos , Lentivirus/metabolismo , Hígado/embriología , Linfocitos/citología , Ratones , Ratones Endogámicos NOD , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Estructura Terciaria de Proteína , Transcripción Genética
12.
Blood ; 118(10): 2712-22, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21791434

RESUMEN

Pleiotrophin (Ptn) is strongly expressed by stromal cells which maintain HSCs. However, in vivo, Ptn deficiency does not alter steady-state hematopoiesis. However, knockdown of Ptn (Ptn(KD)) in stromal cells increases production of hematopoietic progenitors as well as HSC activity in cocultures, suggesting that Ptn may have a role in HSC activation. Indeed, transplantations of wild-type (Ptn(+/+)) HSCs into Ptn(-/-) mice show increased donor cell production in serial transplantations and dominant myeloid regeneration caused by Ptn-dependent regulation of HSC repopulation behavior. This regulation of Lin(-)Kit(+)Sca1(+) function is associated with increased proliferation and, on a molecular level, with up-regulated expression of cyclin D1 (Ccnd1) and C/EBPα (Cepba), but reduced of PPARγ. The known HSC regulator ß-catenin is, however, not altered in the absence of Ptn. In conclusion, our results point to different Ptn-mediated regulatory mechanisms in normal hemostasis and in hematopoietic regeneration and in maintaining the balance of myeloid and lymphoid regeneration. Moreover, our results support the idea that microenvironmental Ptn regulates hematopoietic regeneration through ß-catenin-independent regulation of Ccnd1 and Cebpa.


Asunto(s)
Proteínas Portadoras/fisiología , Proliferación Celular , Citocinas/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células del Estroma/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Western Blotting , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , ARN Mensajero/genética , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , beta Catenina/genética , beta Catenina/metabolismo
14.
iScience ; 26(8): 107328, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37520699

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) describes the age-related acquisition of somatic mutations in hematopoietic stem/progenitor cells (HSPC) leading to clonal blood cell expansion. Although CHIP mutations drive myeloid malignancies like myelodysplastic syndromes (MDS) it is unknown if clonal expansion is attributable to changes in cell type kinetics, or involves reorganization of the hematopoietic hierarchy. Using computational modeling we analyzed differentiation and proliferation kinetics of cultured hematopoietic stem cells (HSC) from 8 healthy individuals, 7 CHIP, and 10 MDS patients. While the standard hematopoietic hierarchy explained HSPC kinetics in healthy samples, 57% of CHIP and 70% of MDS samples were best described with alternative hierarchies. Deregulated kinetics were found at various HSPC compartments with high inter-individual heterogeneity in CHIP and MDS, while altered HSC rates were most relevant in MDS. Quantifying kinetic heterogeneity in detail, we show that reorganization of the HSPC compartment is already detectable in the premalignant CHIP state.

15.
Exp Hematol ; 108: 26-35, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35181392

RESUMEN

GATA2 zinc-finger (ZF) mutations are associated with distinct entities of myeloid malignancies. The specific distribution of these mutations points toward different mechanisms of leukemogenesis depending on the ZF domain affected. In this study, we compared recurring somatic mutations in ZF1 and ZF2. All tested ZF mutants disrupted DNA binding in vitro. In transcription assays, co-expression of FOG1 counteracted GATA2-dependent transcriptional activation, while a variable response to FOG1-mediated repression was observed for individual GATA2 mutants. In primary murine bone marrow cells, GATA2 wild-type (WT) expression inhibited colony formation, while this effect was reduced for both mutants A318T (ZF1) and L359V (ZF2) with a shift toward granulopoiesis. In primary human CD34+ bone marrow cells and in the myeloid cell line K562, ectopic expression of GATA2 L359V, but not A318T or G320D, caused a block of erythroid differentiation accompanied by downregulation of GATA1, STAT5B, and PLCG1. Our findings may explain the role of GATA2 L359V during the progression of chronic myeloid leukemia and the collaboration of GATA2 ZF1 alterations with CEBPA double mutations in erythroleukemia.


Asunto(s)
Factor de Transcripción GATA2 , Leucemia Eritroblástica Aguda , Leucemia Mieloide , Animales , Diferenciación Celular/genética , Factor de Transcripción GATA2/genética , Humanos , Células K562 , Leucemia Eritroblástica Aguda/genética , Ratones , Mutación , Dedos de Zinc
16.
Front Oncol ; 12: 949261, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36263223

RESUMEN

Cellular crosstalk between hematopoietic stem/progenitor cells and the bone marrow (BM) niche is vital for the development and maintenance of myeloid malignancies. These compartments can communicate via bidirectional transfer of extracellular vesicles (EVs). EV trafficking in acute myeloid leukemia (AML) plays a crucial role in shaping the BM microenvironment into a leukemia-permissive niche. Although several EV isolation methods have been developed, it remains a major challenge to define the most accurate and reliable procedure. Here, we tested the efficacy and functional assay compatibility of four different EV isolation methods in leukemia-derived EVs: (1) membrane affinity-based: exoEasy Kit alone and (2) in combination with Amicon filtration; (3) precipitation: ExoQuick-TC; and (4) ultracentrifugation (UC). Western blot analysis of EV fractions showed the highest enrichment of EV marker expression (e.g., CD63, HSP70, and TSG101) by precipitation with removal of overabundant soluble proteins [e.g., bovine serum albumin (BSA)], which were not discarded using UC. Besides the presence of damaged EVs after UC, intact EVs were successfully isolated with all methods as evidenced by highly maintained spherical- and cup-shaped vesicles in transmission electron microscopy. Nanoparticle tracking analysis of EV particle size and concentration revealed significant differences in EV isolation efficacy, with exoEasy Kit providing the highest EV yield recovery. Of note, functional assays with exoEasy Kit-isolated EVs showed significant toxicity towards treated target cells [e.g., mesenchymal stromal cells (MSCs)], which was abrogated when combining exoEasy Kit with Amicon filtration. Additionally, MSC treated with green fluorescent protein (GFP)-tagged exoEasy Kit-isolated EVs did not show any EV uptake, while EV isolation by precipitation demonstrated efficient EV internalization. Taken together, the choice of EV isolation procedure significantly impacts the yield and potential functionality of leukemia-derived EVs. The cheapest method (UC) resulted in contaminated and destructed EV fractions, while the isolation method with the highest EV yield (exoEasy Kit) appeared to be incompatible with functional assays. We identified two methods (precipitation-based ExoQuick-TC and membrane affinity-based exoEasy Kit combined with Amicon filtration) yielding pure and intact EVs, also suitable for application in functional assays. This study highlights the importance of selecting the right EV isolation method depending on the desired experimental design.

17.
Nat Commun ; 13(1): 3456, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35705536

RESUMEN

Plasmacytoid and conventional dendritic cells (pDC and cDC) are generated from progenitor cells in the bone marrow and commitment to pDCs or cDC subtypes may occur in earlier and later progenitor stages. Cells within the CD11c+MHCII-/loSiglec-H+CCR9lo DC precursor fraction of the mouse bone marrow generate both pDCs and cDCs. Here we investigate the heterogeneity and commitment of subsets in this compartment by single-cell transcriptomics and high-dimensional flow cytometry combined with cell fate analysis: Within the CD11c+MHCII-/loSiglec-H+CCR9lo DC precursor pool cells expressing high levels of Ly6D and lacking expression of transcription factor Zbtb46 contain CCR9loB220hi immediate pDC precursors and CCR9loB220lo (lo-lo) cells which still generate pDCs and cDCs in vitro and in vivo under steady state conditions. cDC-primed cells within the Ly6DhiZbtb46- lo-lo precursors rapidly upregulate Zbtb46 and pass through a Zbtb46+Ly6D+ intermediate stage before acquiring cDC phenotype after cell division. Type I IFN stimulation limits cDC and promotes pDC output from this precursor fraction by arresting cDC-primed cells in the Zbtb46+Ly6D+ stage preventing their expansion and differentiation into cDCs. Modulation of pDC versus cDC output from precursors by external factors may allow for adaptation of DC subset composition at later differentiation stages.


Asunto(s)
Antígenos Ly , Células Dendríticas , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Antígeno CD11c/metabolismo , Diferenciación Celular/genética , Células Dendríticas/metabolismo , Proteínas Ligadas a GPI/metabolismo , Ratones , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Células Madre/metabolismo , Factores de Transcripción
18.
Commun Biol ; 5(1): 1246, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36380073

RESUMEN

Stromal cells interact with immune cells during initiation and resolution of immune responses, though the precise underlying mechanisms remain to be resolved. Lessons learned from stromal cell-based therapies indicate that environmental signals instruct their immunomodulatory action contributing to immune response control. Here, to the best of our knowledge, we show a novel function for the guanine-exchange factor DOCK2 in regulating immunosuppressive function in three human stromal cell models and by siRNA-mediated DOCK2 knockdown. To identify immune function-related stromal cell molecular signatures, we first reprogrammed mesenchymal stem/progenitor cells (MSPCs) into induced pluripotent stem cells (iPSCs) before differentiating these iPSCs in a back-loop into MSPCs. The iPSCs and immature iPS-MSPCs lacked immunosuppressive potential. Successive maturation facilitated immunomodulation, while maintaining clonogenicity, comparable to their parental MSPCs. Sequential transcriptomics and methylomics displayed time-dependent immune-related gene expression trajectories, including DOCK2, eventually resembling parental MSPCs. Severe combined immunodeficiency (SCID) patient-derived fibroblasts harboring bi-allelic DOCK2 mutations showed significantly reduced immunomodulatory capacity compared to non-mutated fibroblasts. Conditional DOCK2 siRNA knockdown in iPS-MSPCs and fibroblasts also immediately reduced immunomodulatory capacity. Conclusively, CRISPR/Cas9-mediated DOCK2 knockout in iPS-MSPCs also resulted in significantly reduced immunomodulation, reduced CDC42 Rho family GTPase activation and blunted filopodia formation. These data identify G protein signaling as key element devising stromal cell immunomodulation.


Asunto(s)
Proteínas Activadoras de GTPasa , Guanina , Humanos , Proteínas Activadoras de GTPasa/genética , ARN Interferente Pequeño , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Inmunidad , Inmunomodulación
19.
Nat Commun ; 13(1): 281, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35022408

RESUMEN

SUMOylation is a post-translational modification of proteins that regulates these proteins' localization, turnover or function. Aberrant SUMOylation is frequently found in cancers but its origin remains elusive. Using a genome-wide transposon mutagenesis screen in a MYC-driven B-cell lymphoma model, we here identify the SUMO isopeptidase (or deconjugase) SENP6 as a tumor suppressor that links unrestricted SUMOylation to tumor development and progression. Notably, SENP6 is recurrently deleted in human lymphomas and SENP6 deficiency results in unrestricted SUMOylation. Mechanistically, SENP6 loss triggers release of DNA repair- and genome maintenance-associated protein complexes from chromatin thereby impairing DNA repair in response to DNA damages and ultimately promoting genomic instability. In line with this hypothesis, SENP6 deficiency drives synthetic lethality to Poly-ADP-Ribose-Polymerase (PARP) inhibition. Together, our results link SENP6 loss to defective genome maintenance and reveal the potential therapeutic application of PARP inhibitors in B-cell lymphoma.


Asunto(s)
Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Mutación , Sumoilación/fisiología , Animales , Biomarcadores de Tumor , Liasas de Carbono-Nitrógeno/genética , Liasas de Carbono-Nitrógeno/metabolismo , Cromatina , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Femenino , Inestabilidad Genómica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Sumoilación/efectos de los fármacos , Sumoilación/genética , Mutaciones Letales Sintéticas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Exp Cell Res ; 316(16): 2609-17, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20599957

RESUMEN

Mesenchymal stem cells (MSC) represent a mixture of different cell types, of which only a minority is therapeutically relevant. Surface markers specifically identifying non-differentiated MSC from their differentiated progeny have not been described in sufficient detail. We here compare the gene expression profile of the in vivo bone-forming bone marrow-derived MSC (BM-MSC) with non-bone-forming umbilical vein stromal cells (UVSC) and other non-MSC. Clustering analysis shows that UVSC are a lineage homogeneous cell population, clearly distinct from MSC, other mesenchymal lineages and hematopoietic cells. We find that 89 transcripts of membrane-associated proteins are represented more in cultured BM-MSC than in UVSC. These include previously identified molecules, but also novel markers like NOTCH3, JAG1, and ITGA11. We show that the latter three molecules are also expressed on fibroblast colony-forming units (CFU-F). Both NOTCH3 and ITGA11, but not JAG1, further enrich for CFU-F when combined with CD146, a known marker of cells with MSC activity in vivo. Differentiation studies show that NOTCH3+ and CD146+ NOTCH3+ cells sorted from cultured BM-MSC are capable of adipogenic and osteogenic progeny, while ITGA11-expressing cells mainly show an osteogenic differentiation profile with limited adipogenic differentiation. Our observations may facilitate the study of lineage relationships in MSC as well as facilitate the development of more homogeneous cell populations for mesenchymal cell therapy.


Asunto(s)
Biomarcadores/metabolismo , Médula Ósea/metabolismo , Linaje de la Célula , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo , Venas Umbilicales/metabolismo , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Genoma Humano , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA