Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Immunol ; 23(4): 605-618, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35352063

RESUMEN

Autonomic nerves control organ function through the sympathetic and parasympathetic branches, which have opposite effects. In the bone marrow, sympathetic (adrenergic) nerves promote hematopoiesis; however, how parasympathetic (cholinergic) signals modulate hematopoiesis is unclear. Here, we show that B lymphocytes are an important source of acetylcholine, a neurotransmitter of the parasympathetic nervous system, which reduced hematopoiesis. Single-cell RNA sequencing identified nine clusters of cells that expressed the cholinergic α7 nicotinic receptor (Chrna7) in the bone marrow stem cell niche, including endothelial and mesenchymal stromal cells (MSCs). Deletion of B cell-derived acetylcholine resulted in the differential expression of various genes, including Cxcl12 in leptin receptor+ (LepR+) stromal cells. Pharmacologic inhibition of acetylcholine signaling increased the systemic supply of inflammatory myeloid cells in mice and humans with cardiovascular disease.


Asunto(s)
Acetilcolina , Hematopoyesis , Animales , Linfocitos B , Colinérgicos , Hematopoyesis/genética , Ratones , Nicho de Células Madre
2.
Cell ; 177(7): 1915-1932.e16, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31130381

RESUMEN

Stroma is a poorly defined non-parenchymal component of virtually every organ with key roles in organ development, homeostasis, and repair. Studies of the bone marrow stroma have defined individual populations in the stem cell niche regulating hematopoietic regeneration and capable of initiating leukemia. Here, we use single-cell RNA sequencing (scRNA-seq) to define a cellular taxonomy of the mouse bone marrow stroma and its perturbation by malignancy. We identified seventeen stromal subsets expressing distinct hematopoietic regulatory genes spanning new fibroblastic and osteoblastic subpopulations including distinct osteoblast differentiation trajectories. Emerging acute myeloid leukemia impaired mesenchymal osteogenic differentiation and reduced regulatory molecules necessary for normal hematopoiesis. These data suggest that tissue stroma responds to malignant cells by disadvantaging normal parenchymal cells. Our taxonomy of the stromal compartment provides a comprehensive bone marrow cell census and experimental support for cancer cell crosstalk with specific stromal elements to impair normal tissue function and thereby enable emergent cancer.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Homeostasis , Leucemia Mieloide Aguda/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Microambiente Tumoral , Animales , Células de la Médula Ósea/patología , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Osteoblastos/patología , Células del Estroma/metabolismo , Células del Estroma/patología
3.
Cell ; 166(4): 894-906, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27518564

RESUMEN

Regulation of stem and progenitor cell populations is critical in the development, maintenance, and regeneration of tissues. Here, we define a novel mechanism by which a niche-secreted RNase, angiogenin (ANG), distinctively alters the functional characteristics of primitive hematopoietic stem/progenitor cells (HSPCs) compared with lineage-committed myeloid-restricted progenitor (MyePro) cells. Specifically, ANG reduces the proliferative capacity of HSPC while simultaneously increasing proliferation of MyePro cells. Mechanistically, ANG induces cell-type-specific RNA-processing events: tRNA-derived stress-induced small RNA (tiRNA) generation in HSPCs and rRNA induction in MyePro cells, leading to respective reduction and increase in protein synthesis. Recombinant ANG protein improves survival of irradiated animals and enhances hematopoietic regeneration of mouse and human HSPCs in transplantation. Thus, ANG plays a non-cell-autonomous role in regulation of hematopoiesis by simultaneously preserving HSPC stemness and promoting MyePro proliferation. These cell-type-specific functions of ANG suggest considerable therapeutic potential.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Ribonucleasa Pancreática/metabolismo , Animales , Proliferación Celular , Hematopoyesis , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , ARN de Transferencia/metabolismo , ARN no Traducido/metabolismo
5.
Circ Res ; 124(9): 1372-1385, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30782088

RESUMEN

RATIONALE: After a stroke, patients frequently experience altered systemic immunity resulting in peripheral immunosuppression and higher susceptibility to infections, which is at least partly attributed to lymphopenia. The mechanisms that profoundly change the systemic leukocyte repertoire after stroke are incompletely understood. Emerging evidence indicates that stroke alters hematopoietic output of the bone marrow. OBJECTIVE: To explore the mechanisms that lead to defects of B lymphopoiesis after ischemic stroke. METHODS AND RESULTS: We here report that ischemic stroke triggers brain-bone marrow communication via hormonal long-range signals that regulate hematopoietic B lineage decisions. Bone marrow fluorescence-activated cell sorter analyses and serial intravital microscopy indicate that transient middle cerebral artery occlusion in mice arrests B-cell development beginning at the pro-B-cell stage. This phenotype was not rescued in Myd88-/- and TLR4-/- mice with disrupted TLR (Toll-like receptor) signaling or after blockage of peripheral sympathetic nerves. Mechanistically, we identified stroke-induced glucocorticoid release as the main instigator of B lymphopoiesis defects. B-cell lineage-specific deletion of the GR (glucocorticoid receptor) in CD19-Cre loxP Nr3c1 mice attenuated lymphocytopenia after transient middle cerebral artery. In 20 patients with acute stroke, increased cortisol levels inversely correlated with blood lymphocyte numbers. CONCLUSIONS: Our data demonstrate that the hypothalamic-pituitary-adrenal axis mediates B lymphopoiesis defects after ischemic stroke.


Asunto(s)
Corticoesteroides/sangre , Linfocitos B/metabolismo , Células de la Médula Ósea/metabolismo , Linfopoyesis , Receptores de Glucocorticoides/sangre , Accidente Cerebrovascular/sangre , Anciano , Animales , Linfocitos B/citología , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Sistema Hipófiso-Suprarrenal/fisiopatología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/fisiopatología
6.
Circ Res ; 116(3): 407-17, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25362208

RESUMEN

RATIONALE: The mechanisms leading to an expanded neutrophil and monocyte supply after stroke are incompletely understood. OBJECTIVE: To test the hypothesis that transient middle cerebral artery occlusion (tMCAO) in mice leads to activation of hematopoietic bone marrow stem cells. METHODS AND RESULTS: Serial in vivo bioluminescence reporter gene imaging in mice with tMCAO revealed that bone marrow cell cycling peaked 4 days after stroke (P<0.05 versus pre tMCAO). Flow cytometry and cell cycle analysis showed activation of the entire hematopoietic tree, including myeloid progenitors. The cycling fraction of the most upstream hematopoietic stem cells increased from 3.34%±0.19% to 7.32%±0.52% after tMCAO (P<0.05). In vivo microscopy corroborated proliferation of adoptively transferred hematopoietic progenitors in the bone marrow of mice with stroke. The hematopoietic system's myeloid bias was reflected by increased expression of myeloid transcription factors, including PU.1 (P<0.05), and by a decline in lymphocyte precursors. In mice after tMCAO, tyrosine hydroxylase levels in sympathetic fibers and bone marrow noradrenaline levels rose (P<0.05, respectively), associated with a decrease of hematopoietic niche factors that promote stem cell quiescence. In mice with genetic deficiency of the ß3 adrenergic receptor, hematopoietic stem cells did not enter the cell cycle in increased numbers after tMCAO (naive control, 3.23±0.22; tMCAO, 3.74±0.33, P=0.51). CONCLUSIONS: Ischemic stroke activates hematopoietic stem cells via increased sympathetic tone, leading to a myeloid bias of hematopoiesis and higher bone marrow output of inflammatory Ly6C(high) monocytes and neutrophils.


Asunto(s)
Infarto de la Arteria Cerebral Media/patología , Células Madre Mesenquimatosas/fisiología , Mielopoyesis , Fibras Adrenérgicas/metabolismo , Fibras Adrenérgicas/fisiología , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Ciclo Celular , Infarto de la Arteria Cerebral Media/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Nicho de Células Madre , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
7.
Stem Cells ; 31(7): 1340-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23533197

RESUMEN

The identification of the molecular mechanisms controlling the degradation of regulatory proteins in mesenchymal stromal cells (MSC) may provide clues to promote MSC osteogenic differentiation and bone regeneration. Ubiquitin ligase-dependent degradation of proteins is an important process governing cell fate. In this study, we investigated the role of the E3 ubiquitin ligase c-Cbl in MSC osteoblast differentiation and identified the mechanisms involved in this effect. Using distinct shRNA targeting c-Cbl, we showed that c-Cbl silencing promotes osteoblast differentiation in murine and human MSC, as demonstrated by increased alkaline phosphatase activity, expression of phenotypic osteoblast marker genes (RUNX2, ALP, type 1 collagen), and matrix mineralization in vitro. Coimmunoprecipitation analyses showed that c-Cbl interacts with the transcription factor STAT5, and that STAT5 forms a complex with RUNX2, a master transcription factor controlling osteoblastogenesis. Silencing c-Cbl decreased c-Cbl-mediated STAT5 ubiquitination, increased STAT5 protein level and phosphorylation, and enhanced STAT5 and RUNX2 transcriptional activity. The expression of insulin like growth factor-1 (IGF-1), a target gene of STAT5, was increased by c-Cbl silencing in MSC and in bone marrow stromal cells isolated from c-Cbl deficient mice, suggesting that IGF-1 contributes to osteoblast differentiation induced by c-Cbl silencing in MSC. Consistent with these findings, pharmacological inhibition of STAT5 activity, or neutralization of IGF-1 activity, abrogated the positive effect of c-Cbl knockdown on MSC osteogenic differentiation. Taken together, the data provide a novel functional mechanism by which the ubiquitin ligase c-Cbl regulates the osteoblastic differentiation program in mesenchymal cells by controlling Cbl-mediated STAT5 degradation and activity.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Factor de Transcripción STAT5/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factor de Transcripción STAT5/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética
8.
J Biol Chem ; 286(27): 24443-50, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21596750

RESUMEN

Human bone marrow-derived mesenchymal stromal cells (hMSCs) have the capacity to differentiate into several cell types including osteoblasts and are therefore an important cell source for bone tissue regeneration. A crucial issue is to identify mechanisms that trigger hMSC osteoblast differentiation to promote osteogenic potential. Casitas B lineage lymphoma (Cbl) is an E3 ubiquitin ligase that ubiquitinates and targets several molecules for degradation. We hypothesized that attenuation of Cbl-mediated degradation of receptor tyrosine kinases (RTKs) may promote osteogenic differentiation in hMSCs. We show here that specific inhibition of Cbl interaction with RTKs using a Cbl mutant (G306E) promotes expression of osteoblast markers (Runx2, alkaline phosphatase, type 1 collagen, osteocalcin) and increases osteogenic differentiation in clonal bone marrow-derived hMSCs and primary hMSCs. Analysis of molecular mechanisms revealed that the Cbl mutant increased PDGF receptor α and FGF receptor 2 but not EGF receptor expression in hMSCs, resulting in increased ERK1/2 and PI3K signaling. Pharmacological inhibition of FGFR or PDGFR abrogated in vitro osteogenesis induced by the Cbl mutant. The data reveal that specific inhibition of Cbl interaction with RTKs promotes the osteogenic differentiation program in hMSCs in part by decreased Cbl-mediated PDGFRα and FGFR2 ubiquitination, providing a novel mechanistic approach targeting Cbl to promote the osteogenic capacity of hMSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Mutación Missense , Proteína Oncogénica v-cbl/metabolismo , Osteogénesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Sustitución de Aminoácidos , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Células de la Médula Ósea/citología , Línea Celular Transformada , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-cbl/antagonistas & inhibidores , Proteína Oncogénica v-cbl/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/citología , Células del Estroma/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética
9.
Growth Factors ; 30(2): 117-23, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22292523

RESUMEN

Fibroblast growth factors (FGFs) are important molecules that control bone formation. FGF act by activating FGF receptors (FGFRs) and downstream signaling pathways that control cells of the osteoblast lineage. Recent advances have been made in the identification of FGF/FGFR signaling pathways that control osteogenesis. Indeed, studies of mouse and human models provided novel insights into the signaling pathways that control bone formation. Genomic studies also highlighted the implication of molecular targets of FGF/FGFR signaling regulating osteoblastogenesis. Recent studies further revealed the important role of crosstalks between FGF/FGFR signaling and other signaling pathways in the regulation of osteogenesis. Finally, the importance of the mechanisms modulating FGFR degradation in the control of osteoblast differentiation has been recently revealed. This short review summarizes the recently described mechanisms underlying FGF/FGFR signaling that are involved in the control of osteoblastogenesis. This knowledge may have potential therapeutic implications in skeletal disorders characterized by abnormal bone formation.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Osteogénesis/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Animales , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Osteoblastos/citología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología
10.
Med Sci (Paris) ; 28(11): 970-5, 2012 Nov.
Artículo en Francés | MEDLINE | ID: mdl-23171901

RESUMEN

Cbl ubiquitin ligases are important molecules that control the process of ubiquitination and degradation of proteins by the proteasome. Because this process regulates several intracellular mechanisms, alterations in Cbl activity lead to several pathologies including cancer. In bone, the c-Cbl ubiquitin ligase is known to control osteoclast activity. Our studies indicate that c-Cbl also regulates osteoblast proliferation, differentiation and survival. We recently showed that inhibition of c-Cbl activity using a c-Cbl mutant leads to promote osteoblast differentiation in mesenchymal stromal cells as a consequence of increased receptor tyrosine kinase expression. Conversely, we found that overexpression of c-Cbl leads to inhibit osteosarcoma cell proliferation and tumorigenesis through downregulation of these receptors. Thus, the use of pharmacological agents capable of modulating c-Cbl activity may be of therapeutic interest for promoting bone formation in normal bone, or to reduce tumorigenesis in primary bone cancer.


Asunto(s)
Neoplasias Óseas/enzimología , Resorción Ósea/enzimología , Transformación Celular Neoplásica , Proteínas de Neoplasias/fisiología , Osteoblastos/enzimología , Osteoclastos/enzimología , Osteogénesis/fisiología , Osteosarcoma/enzimología , Proteínas Proto-Oncogénicas c-cbl/fisiología , Animales , Neoplasias Óseas/tratamiento farmacológico , Diferenciación Celular , División Celular , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-cbl/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/fisiología , Ubiquitinación
11.
Cancer Cell ; 39(11): 1464-1478.e8, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34719426

RESUMEN

Bone metastases are devastating complications of cancer. They are particularly common in prostate cancer (PCa), represent incurable disease, and are refractory to immunotherapy. We seek to define distinct features of the bone marrow (BM) microenvironment by analyzing single cells from bone metastatic prostate tumors, involved BM, uninvolved BM, and BM from cancer-free, orthopedic patients, and healthy individuals. Metastatic PCa is associated with multifaceted immune distortion, specifically exhaustion of distinct T cell subsets, appearance of macrophages with states specific to PCa bone metastases. The chemokine CCL20 is notably overexpressed by myeloid cells, as is its cognate CCR6 receptor on T cells. Disruption of the CCL20-CCR6 axis in mice with syngeneic PCa bone metastases restores T cell reactivity and significantly prolongs animal survival. Comparative high-resolution analysis of PCa bone metastases shows a targeted approach for relieving local immunosuppression for therapeutic effect.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Quimiocina CCL20/genética , Neoplasias de la Próstata/patología , Receptores CCR6/genética , Regulación hacia Arriba , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/inmunología , Estudios de Casos y Controles , Línea Celular Tumoral , Quimiocina CCL20/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Macrófagos/inmunología , Masculino , Ratones , Células Mieloides/inmunología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Receptores CCR6/metabolismo , Análisis de la Célula Individual , Linfocitos T/inmunología , Microambiente Tumoral
12.
Cell Stem Cell ; 28(12): 2090-2103.e9, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34551362

RESUMEN

Extracellular vesicles (EVs) transfer complex biologic material between cells. However, the role of this process in vivo is poorly defined. Here, we demonstrate that osteoblastic cells in the bone marrow (BM) niche elaborate extracellular vesicles that are taken up by hematopoietic progenitor cells in vivo. Genotoxic or infectious stress rapidly increased stromal-derived extracellular vesicle transfer to granulocyte-monocyte progenitors. The extracellular vesicles contained processed tRNAs (tiRNAs) known to modulate protein translation. 5'-ti-Pro-CGG-1 was preferentially abundant in osteoblast-derived extracellular vesicles and, when transferred to granulocyte-monocyte progenitors, increased protein translation, cell proliferation, and myeloid differentiation. Upregulating EV transfer improved hematopoietic recovery from genotoxic injury and survival from fungal sepsis. Therefore, EV-mediated tiRNA transfer provides a stress-modulated signaling axis in the BM niche distinct from conventional cytokine-driven stress responses.


Asunto(s)
Vesículas Extracelulares , Células Madre Hematopoyéticas , Médula Ósea , Células de la Médula Ósea , Hematopoyesis
13.
J Cell Biochem ; 110(5): 1147-54, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564211

RESUMEN

The capacity of mesenchymal stem cells (MSCs) to differentiate into functional osteoblasts is tightly controlled by transcription factors that trigger osteoblast commitment and differentiation. The role of Twist1, a basic helix-loop-helix (bHLH) transcription factor, in osteogenic differentiation of MSCs remains unclear. Here we investigated the role of Twist1 in the osteogenic differentiation program of murine C3H10T1/2 mesenchymal cells. We showed that molecular silencing of Twist1 using short hairpin RNA (shRNA) expression moderately increased C3H10T1/2 cell proliferation and had no effect on cell survival. In contrast, Twist1 silencing enhanced osteoblast gene expression and matrix mineralization in vitro. Biochemical analyses revealed that Twist1 silencing increased the expression of FGFR2 protein level, which was reduced by a mutant Runx2. Consistent with this finding, Twist1 silencing increased ERK1/2 and PI3K signaling. Moreover, molecular or pharmacological inhibition of FGFR2 or of ERK1/2 and PI3K signaling partly abolished the increased osteoblast gene expression induced by Twist1 silencing in C3H10T1/2 cells. These results reveal that Twist1 silencing upregulates osteoblast differentiation of murine mesenchymal cells in part via activation of FGFR2 expression and downstream signaling pathways, which provides novel insights into the molecular signals by which this transcription factor regulates the osteogenic differentiation program in MSCs.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Interferencia de ARN , Proteína 1 Relacionada con Twist/metabolismo , Animales , Western Blotting , Línea Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C3H , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Nucleares/genética , Osteoblastos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteína 1 Relacionada con Twist/genética
14.
Cell Stem Cell ; 25(4): 570-583.e7, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31279774

RESUMEN

Stromal cell populations that maintain hematopoietic stem and progenitor cells (HSPCs) are generally characterized in steady-state conditions. Here, we report a comprehensive atlas of bone marrow stromal cell subpopulations under homeostatic and stress conditions using mass cytometry (CyTOF)-based single-cell protein analysis. We identified 28 subsets of non-hematopoietic cells during homeostasis, 14 of which expressed hematopoietic regulatory factors. Irradiation-based conditioning for HSPC transplantation led to the loss of most of these populations, including the LeptinR+ and Nestin+ subsets. In contrast, a subset expressing Ecto-5'-nucleotidase (CD73) was retained and a specific CD73+NGFRhigh population expresses high levels of cytokines during homeostasis and stress. Genetic ablation of CD73 compromised HSPC transplantation in an acute setting without long-term changes in bone marrow HSPCs. Thus, this protein-based expression mapping reveals distinct sets of stromal cells in the bone marrow and how they change in clinically relevant stress settings to contribute to early stages of hematopoietic regeneration.


Asunto(s)
Células de la Médula Ósea/metabolismo , Estrés Fisiológico/fisiología , Células del Estroma/metabolismo , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Animales , Atlas como Asunto , Células de la Médula Ósea/patología , Células Cultivadas , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Homeostasis , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Nestina/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptores de Leptina/metabolismo , Nicho de Células Madre , Células del Estroma/patología
15.
Nat Med ; 25(11): 1761-1771, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31700184

RESUMEN

A sedentary lifestyle, chronic inflammation and leukocytosis increase atherosclerosis; however, it remains unclear whether regular physical activity influences leukocyte production. Here we show that voluntary running decreases hematopoietic activity in mice. Exercise protects mice and humans with atherosclerosis from chronic leukocytosis but does not compromise emergency hematopoiesis in mice. Mechanistically, exercise diminishes leptin production in adipose tissue, augmenting quiescence-promoting hematopoietic niche factors in leptin-receptor-positive stromal bone marrow cells. Induced deletion of the leptin receptor in Prrx1-creERT2; Leprfl/fl mice reveals that leptin's effect on bone marrow niche cells regulates hematopoietic stem and progenitor cell (HSPC) proliferation and leukocyte production, as well as cardiovascular inflammation and outcomes. Whereas running wheel withdrawal quickly reverses leptin levels, the impact of exercise on leukocyte production and on the HSPC epigenome and transcriptome persists for several weeks. Together, these data show that physical activity alters HSPCs via modulation of their niche, reducing hematopoietic output of inflammatory leukocytes.


Asunto(s)
Aterosclerosis/terapia , Enfermedades Cardiovasculares/terapia , Células Madre Hematopoyéticas/metabolismo , Inflamación/terapia , Condicionamiento Físico Animal , Tejido Adiposo/metabolismo , Animales , Aterosclerosis/prevención & control , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Epigenoma/genética , Ejercicio Físico/fisiología , Hematopoyesis/genética , Hematopoyesis/fisiología , Proteínas de Homeodominio/genética , Humanos , Inflamación/fisiopatología , Leucocitos/metabolismo , Leucocitosis/fisiopatología , Leucocitosis/terapia , Ratones , Receptores de Leptina/genética , Conducta Sedentaria , Transcriptoma/genética
16.
Genome Biol ; 19(1): 78, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29921301

RESUMEN

Recent single-cell RNA-seq protocols based on droplet microfluidics use massively multiplexed barcoding to enable simultaneous measurements of transcriptomes for thousands of individual cells. The increasing complexity of such data creates challenges for subsequent computational processing and troubleshooting of these experiments, with few software options currently available. Here, we describe a flexible pipeline for processing droplet-based transcriptome data that implements barcode corrections, classification of cell quality, and diagnostic information about the droplet libraries. We introduce advanced methods for correcting composition bias and sequencing errors affecting cellular and molecular barcodes to provide more accurate estimates of molecular counts in individual cells.


Asunto(s)
Código de Barras del ADN Taxonómico/métodos , ARN/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Células K562 , Masculino , Ratones , Ratones Endogámicos C57BL , Microfluídica/métodos , Programas Informáticos , Transcriptoma/genética
17.
J Clin Invest ; 128(1): 281-293, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202481

RESUMEN

Nervous system injury is a frequent result of cancer therapy involving cranial irradiation, leaving patients with marked memory and other neurobehavioral disabilities. Here, we report an unanticipated link between bone marrow and brain in the setting of radiation injury. Specifically, we demonstrate that bone marrow-derived monocytes and macrophages are essential for structural and functional repair mechanisms, including regeneration of cerebral white matter and improvement in neurocognitive function. Using a granulocyte-colony stimulating factor (G-CSF) receptor knockout mouse model in combination with bone marrow cell transplantation, MRI, and neurocognitive functional assessments, we demonstrate that bone marrow-derived G-CSF-responsive cells home to the injured brain and are critical for altering neural progenitor cells and brain repair. Additionally, compared with untreated animals, animals that received G-CSF following radiation injury exhibited enhanced functional brain repair. Together, these results demonstrate that, in addition to its known role in defense and debris removal, the hematopoietic system provides critical regenerative drive to the brain that can be modulated by clinically available agents.


Asunto(s)
Células de la Médula Ósea , Trasplante de Médula Ósea , Encéfalo/fisiología , Factor Estimulante de Colonias de Granulocitos/farmacología , Trastornos Neurocognitivos , Traumatismos Experimentales por Radiación , Regeneración/efectos de los fármacos , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Trastornos Neurocognitivos/genética , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/fisiopatología , Trastornos Neurocognitivos/terapia , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/fisiopatología , Traumatismos Experimentales por Radiación/terapia , Regeneración/genética , Regeneración/efectos de la radiación
18.
Cancer Res ; 78(18): 5300-5314, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30065048

RESUMEN

The presence of disseminated tumor cells in breast cancer patient bone marrow aspirates predicts decreased recurrence-free survival. Although it is appreciated that physiologic, pathologic, and therapeutic conditions impact hematopoiesis, it remains unclear whether targeting hematopoiesis presents opportunities for limiting bone metastasis. Using preclinical breast cancer models, we discovered that marrow from mice treated with the bisphosphonate zoledronic acid (ZA) are metastasis-suppressive. Specifically, ZA modulated hematopoietic myeloid/osteoclast progenitor cell (M/OCP) lineage potential to activate metastasis-suppressive activity. Granulocyte-colony stimulating factor (G-CSF) promoted ZA resistance by redirecting M/OCP differentiation. We identified M/OCP and bone marrow transcriptional programs associated with metastasis suppression and ZA resistance. Analysis of patient blood samples taken at randomization revealed that women with high-plasma G-CSF experienced significantly worse outcome with adjuvant ZA than those with lower G-CSF levels. Our findings support discovery of therapeutic strategies to direct M/OCP lineage potential and biomarkers that stratify responses in patients at risk of recurrence.Significance: Bone marrow myeloid/osteoclast progenitor cell lineage potential has a profound impact on breast cancer bone metastasis and can be modulated by G-CSF and bone-targeting agents. Cancer Res; 78(18); 5300-14. ©2018 AACR.


Asunto(s)
Células de la Médula Ósea/citología , Neoplasias de la Mama/patología , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Metástasis de la Neoplasia/prevención & control , Animales , Antineoplásicos/farmacología , Biomarcadores/metabolismo , Médula Ósea/patología , Neoplasias Óseas/prevención & control , Diferenciación Celular , Línea Celular Tumoral , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Hematopoyesis , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Recurrencia Local de Neoplasia , Osteoclastos/citología , Osteoclastos/metabolismo , Ácido Zoledrónico/farmacología
19.
Cell Stem Cell ; 20(5): 590-592, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475884

RESUMEN

The niche, as first conceptualized, was in conflict with the prevailing wisdom that stem cells have internal logic. The niche hypothesis has been indisputably confirmed. Yet, recent findings indicate little plasticity of epigenetically scripted hematopoietic stem/progenitors. Reconciling this conflict requires re-envisioning the niche as an enabler, not designer, of cell fate.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Ratones , ARN Mensajero/genética , Nicho de Células Madre/genética
20.
Science ; 358(6367)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191879

RESUMEN

Bone marrow-derived myeloid cells can accumulate within tumors and foster cancer outgrowth. Local immune-neoplastic interactions have been intensively investigated, but the contribution of the systemic host environment to tumor growth remains poorly understood. Here, we show in mice and cancer patients (n = 70) that lung adenocarcinomas increase bone stromal activity in the absence of bone metastasis. Animal studies reveal that the cancer-induced bone phenotype involves bone-resident osteocalcin-expressing (Ocn+) osteoblastic cells. These cells promote cancer by remotely supplying a distinct subset of tumor-infiltrating SiglecFhigh neutrophils, which exhibit cancer-promoting properties. Experimentally reducing Ocn+ cell numbers suppresses the neutrophil response and lung tumor outgrowth. These observations posit osteoblasts as remote regulators of lung cancer and identify SiglecFhigh neutrophils as myeloid cell effectors of the osteoblast-driven protumoral response.


Asunto(s)
Adenocarcinoma/patología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Huesos/patología , Lectinas/metabolismo , Neoplasias Pulmonares/patología , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Osteoblastos/patología , Adenocarcinoma del Pulmón , Animales , Densidad Ósea , Células de la Médula Ósea/patología , Huesos/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Neoplasias Experimentales/patología , Osteocalcina/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA