Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Blood ; 136(7): 885-897, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32294156

RESUMEN

Idiopathic aplastic anemia (AA) has 2 key characteristics: an autoimmune response against hematopoietic stem/progenitor cells and regulatory T-cells (Tregs) deficiency. We have previously demonstrated reduction in a specific subpopulation of Treg in AA, which predicts response to immunosuppression. The aims of the present study were to define mechanisms of Treg subpopulation imbalance and identify potential for therapeutic intervention. We have identified 2 mechanisms that lead to skewed Treg composition in AA: first, FasL-mediated apoptosis on ligand interaction; and, second, relative interleukin-2 (IL-2) deprivation. We have shown that IL-2 augmentation can overcome these mechanisms. Interestingly, when high concentrations of IL-2 were used for in vitro Treg expansion cultures, AA Tregs were able to expand. The expanded populations expressed a high level of p-BCL-2, which makes them resistant to apoptosis. Using a xenograft mouse model, the function and stability of expanded AA Tregs were tested. We have shown that these Tregs were able to suppress the macroscopic clinical features and tissue manifestations of T-cell-mediated graft-versus-host disease. These Tregs maintained their suppressive properties as well as their phenotype in a highly inflammatory environment. Our findings provide an insight into the mechanisms of Treg reduction in AA. We have identified novel targets with potential for therapeutic interventions. Supplementation of ex vivo expansion cultures of Tregs with high concentrations of IL-2 or delivery of IL-2 directly to patients could improve clinical outcomes in addition to standard immunosuppressive therapy.


Asunto(s)
Anemia Aplásica/inmunología , Apoptosis/efectos de los fármacos , Proteína Ligando Fas/farmacología , Interleucina-2/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Anemia Aplásica/patología , Animales , Apoptosis/inmunología , Células Cultivadas , Femenino , Humanos , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/patología , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Interleucina-2/deficiencia , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Linfocitos T Reguladores/fisiología
2.
Dev Dyn ; 249(3): 354-368, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31408233

RESUMEN

BACKGROUND: The basic ground plan of vertebrate hindbrain is established through a process of segmentation, which generates eight transient lineage-restricted cellular compartments called rhombomeres (r). The segments adopt distinct individual identities in response to axial patterning signals. It is unclear whether signaling between rhombomeres plays a conserved role in regulating segmental patterning during hindbrain development. RESULTS: Using tissue manipulations of rhombomeres in chicken embryos, we have uncovered roles for r2 and r4 in regulating the expression of EphA4 in r3 and r5. Perturbations of signaling pathways reveal that these regulatory inputs from r2 and r4 into EphA4 expression are mediated independent of inputs from Krox20 through cues involving fibroblast growth factor (FGF) signaling. These interactions are stage dependent and are set up in embryos with <10 somites. CONCLUSIONS: We show that r2 and r4 function as temporally dynamic signaling centers in the early patterning of adjacent hindbrain segments and this activity is dependent upon the FGF pathway. These results reveal that inter-rhombomeric signaling is a conserved feature of the regulatory networks that control the specification of individual rhombomere identities in vertebrate hindbrain segmentation. However, the timing of when restricted domains of FGF signaling are coupled to formation of r4 may vary between the species.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Receptor EphA4/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Rombencéfalo/metabolismo , Animales , Embrión de Pollo , Factores de Crecimiento de Fibroblastos/genética , Hibridación in Situ , Receptor EphA4/genética , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Rombencéfalo/embriología
3.
Blood ; 125(26): 4060-8, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-25896651

RESUMEN

The strategy of enzymatic degradation of amino acids to deprive malignant cells of important nutrients is an established component of induction therapy of acute lymphoblastic leukemia. Here we show that acute myeloid leukemia (AML) cells from most patients with AML are deficient in a critical enzyme required for arginine synthesis, argininosuccinate synthetase-1 (ASS1). Thus, these ASS1-deficient AML cells are dependent on importing extracellular arginine. We therefore investigated the effect of plasma arginine deprivation using pegylated arginine deiminase (ADI-PEG 20) against primary AMLs in a xenograft model and in vitro. ADI-PEG 20 alone induced responses in 19 of 38 AMLs in vitro and 3 of 6 AMLs in vivo, leading to caspase activation in sensitive AMLs. ADI-PEG 20-resistant AMLs showed higher relative expression of ASS1 than sensitive AMLs. This suggests that the resistant AMLs survive by producing arginine through this metabolic pathway and ASS1 expression could be used as a biomarker for response. Sensitive AMLs showed more avid uptake of arginine from the extracellular environment consistent with their auxotrophy for arginine. The combination of ADI-PEG 20 and cytarabine chemotherapy was more effective than either treatment alone resulting in responses in 6 of 6 AMLs tested in vivo. Our data show that arginine deprivation is a reasonable strategy in AML that paves the way for clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Hidrolasas/farmacología , Leucemia Mieloide Aguda/metabolismo , Polietilenglicoles/farmacología , Animales , Arginina/metabolismo , Argininosuccinato Sintasa/biosíntesis , Argininosuccinato Sintasa/genética , Western Blotting , Células Cultivadas , Cromatografía Líquida de Alta Presión , Humanos , Inmunohistoquímica , Leucemia Mieloide Aguda/genética , Espectrometría de Masas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Reacción en Cadena en Tiempo Real de la Polimerasa , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Development ; 138(14): 2947-56, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21653612

RESUMEN

We describe the production and characterisation of two monoclonal antibodies, zdc2 and zdd2, directed against the zebrafish Notch ligands DeltaC and DeltaD, respectively. We use our antibodies to show that these Delta proteins can bind to one another homo- and heterophilically, and to study the localisation of DeltaC and DeltaD in the zebrafish nervous system and presomitic mesoderm (PSM). Our findings in the nervous system largely confirm expectations from previous studies, but in the PSM we see an unexpected pattern in which the localisation of DeltaD varies according to the level of expression of DeltaC: in the anterior PSM, where DeltaC is plentiful, the two proteins are colocalised in intracellular puncta, but in the posterior PSM, where DeltaC is at a lower level, DeltaD is seen mainly on the cell surface. Forced overexpression of DeltaC reduces the amount of DeltaD on the cell surface in the posterior PSM; conversely, loss-of-function mutation of DeltaC increases the amount of DeltaD on the cell surface in the anterior PSM. These findings suggest an explanation for a long-standing puzzle regarding the functions of the two Delta proteins in the somite segmentation clock--an explanation that is based on the proposition that they associate heterophilically to activate Notch.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de la Membrana/inmunología , Mesodermo/metabolismo , Microscopía Confocal , Proteínas del Tejido Nervioso/inmunología , Sistema Nervioso/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas de Pez Cebra/inmunología
5.
Hemasphere ; 8(5): e80, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774656

RESUMEN

Immunodeficient mouse models are widely used for the assessment of human normal and leukemic stem cells. Despite the advancements over the years, reproducibility, as well as the differences in the engraftment of human cells in recipient mice remains to be fully resolved. Here, we used various immunodeficient mouse models to characterize the effect of donor-recipient sex on the engraftment of the human leukemic and healthy cells. Donor human cells and recipient immunodeficient mice demonstrate sex-specific engraftment levels with significant differences observed in the lineage output of normal CD34+ hematopoietic stem and progenitor cells upon xenotransplantation. Intriguingly, human female donor cells display heightened sensitivity to the recipient mice's gender, influencing their proliferation and resulting in significantly increased engraftment in female recipient mice. Our study underscores the intricate interplay taking place between donor and recipient characteristics, shedding light on important considerations for future studies, particularly in the context of pre-clinical research.

6.
Hemasphere ; 7(3): e853, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36874381

RESUMEN

Long-term hematopoietic stem cells are rare, highly quiescent stem cells of the hematopoietic system with life-long self-renewal potential and the ability to transplant and reconstitute the entire hematopoietic system of conditioned recipients. Most of our understanding of these rare cells has relied on cell surface identification, epigenetic, and transcriptomic analyses. Our knowledge of protein synthesis, folding, modification, and degradation-broadly termed protein homeostasis or "proteostasis"-in these cells is still in its infancy, with very little known about how the functional state of the proteome is maintained in hematopoietic stem cells. We investigated the requirement of the small phospho-binding adaptor proteins, the cyclin-dependent kinase subunits (CKS1 and CKS2), for maintaining ordered hematopoiesis and long-term hematopoietic stem cell reconstitution. CKS1 and CKS2 are best known for their roles in p27 degradation and cell cycle regulation, and by studying the transcriptome and proteome of Cks1 -/- and Cks2 -/- mice, we demonstrate regulation of key signaling pathways that govern hematopoietic stem cell biology including AKT, FOXO1, and NFκB, together balancing protein homeostasis and restraining reactive oxygen species to ensure healthy hematopoietic stem cell function.

7.
Dev Cell ; 58(22): 2428-2446.e9, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37652013

RESUMEN

Thymus is necessary for lifelong immunological tolerance and immunity. It displays a distinctive epithelial complexity and undergoes age-dependent atrophy. Nonetheless, it also retains regenerative capacity, which, if harnessed appropriately, might permit rejuvenation of adaptive immunity. By characterizing cortical and medullary compartments in the human thymus at single-cell resolution, in this study we have defined specific epithelial populations, including those that share properties with bona fide stem cells (SCs) of lifelong regenerating epidermis. Thymic epithelial SCs display a distinctive transcriptional profile and phenotypic traits, including pleiotropic multilineage potency, to give rise to several cell types that were not previously considered to have shared origin. Using here identified SC markers, we have defined their cortical and medullary niches and shown that, in vitro, the cells display long-term clonal expansion and self-organizing capacity. These data substantively broaden our knowledge of SC biology and set a stage for tackling thymic atrophy and related disorders.


Asunto(s)
Células Madre , Timo , Humanos , Diferenciación Celular , Células Madre/metabolismo , Timo/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Atrofia/metabolismo
8.
Cell Stem Cell ; 30(6): 781-799.e9, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267914

RESUMEN

Somatic mutations commonly occur in hematopoietic stem cells (HSCs). Some mutant clones outgrow through clonal hematopoiesis (CH) and produce mutated immune progenies shaping host immunity. Individuals with CH are asymptomatic but have an increased risk of developing leukemia, cardiovascular and pulmonary inflammatory diseases, and severe infections. Using genetic engineering of human HSCs (hHSCs) and transplantation in immunodeficient mice, we describe how a commonly mutated gene in CH, TET2, affects human neutrophil development and function. TET2 loss in hHSCs produce a distinct neutrophil heterogeneity in bone marrow and peripheral tissues by increasing the repopulating capacity of neutrophil progenitors and giving rise to low-granule neutrophils. Human neutrophils that inherited TET2 mutations mount exacerbated inflammatory responses and have more condensed chromatin, which correlates with compact neutrophil extracellular trap (NET) production. We expose here physiological abnormalities that may inform future strategies to detect TET2-CH and prevent NET-mediated pathologies associated with CH.


Asunto(s)
Dioxigenasas , Neutrófilos , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas , Células Madre Hematopoyéticas/fisiología , Médula Ósea , Hematopoyesis/genética , Mutación , Proteínas de Unión al ADN/genética , Dioxigenasas/genética
9.
Nat Commun ; 13(1): 2048, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440586

RESUMEN

The heterogeneous nature of human CD34+ hematopoietic stem cells (HSCs) has hampered our understanding of the cellular and molecular trajectories that HSCs navigate during lineage commitment. Using various platforms including single cell RNA-sequencing and extensive xenotransplantation, we have uncovered an uncharacterized human CD34+ HSC population. These CD34+EPCR+(CD38/CD45RA)- (simply as EPCR+) HSCs have a high repopulating and self-renewal abilities, reaching a stem cell frequency of ~1 in 3 cells, the highest described to date. Their unique transcriptomic wiring in which many gene modules associated with differentiated cell lineages confers their multilineage lineage output both in vivo and in vitro. At the single cell level, EPCR+ HSCs are the most transcriptomically and functionally homogenous human HSC population defined to date and can also be easily identified in post-natal tissues. Therefore, this EPCR+ population not only offers a high human HSC resolution but also a well-structured human hematopoietic hierarchical organization at the most primitive level.


Asunto(s)
Células Madre Hematopoyéticas , Análisis de la Célula Individual , Antígenos CD34 , Moléculas de Adhesión Celular , Linaje de la Célula , Receptor de Proteína C Endotelial , Humanos
11.
Blood Cancer Discov ; 2(2): 135-145, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33778768

RESUMEN

Myelodysplastic syndrome (MDS) are clonal stem cell diseases characterized mainly by ineffective hematopoiesis. Here, we present an approach that enables robust long-term engraftment of primary MDS stem cells (MDS-SCs) in mice by implantation of human mesenchymal cell-seeded scaffolds. Critically for modelling MDS, where patient sample material is limiting, mononuclear bone marrow cells containing as few as 104 CD34+ cells can be engrafted and expanded by this approach with the maintenance of the genetic make-up seen in the patients. Non-invasive high-resolution ultrasound imaging shows that these scaffolds are fully perfused. Our data shows that human microenvironment but not mouse is essential to MDS-SCs homing and engraftment. Notably, the alternative niche provided by healthy donor MSCs enhanced engraftment of MDS-SCs. This study characterizes a new tool to model MDS human disease with the level of engraftment previously unattainable in mice, and offers insights into human-specific determinants of MDS-SC microenvironment.


Asunto(s)
Células Madre Mesenquimatosas , Síndromes Mielodisplásicos , Animales , Células de la Médula Ósea , Hematopoyesis , Humanos , Ratones , Células Madre
12.
Cell Rep ; 35(6): 109119, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979628

RESUMEN

The bone-marrow (BM) niche is the spatial environment composed by a network of multiple stromal components regulating adult hematopoiesis. We use multi-omics and computational tools to analyze multiple BM environmental compartments and decipher their mutual interactions in the context of acute myeloid leukemia (AML) xenografts. Under homeostatic conditions, we find a considerable overlap between niche populations identified using current markers. Our analysis defines eight functional clusters of genes informing on the cellular identity and function of the different subpopulations and pointing at specific stromal interrelationships. We describe how these transcriptomic profiles change during human AML development and, by using a proximity-based molecular approach, we identify early disease onset deregulated genes in the mesenchymal compartment. Finally, we analyze the BM proteomic secretome in the presence of AML and integrate it with the transcriptome to predict signaling nodes involved in niche alteration in AML.


Asunto(s)
Células de la Médula Ósea/metabolismo , Leucemia Mieloide Aguda/genética , Proteómica/métodos , Animales , Humanos , Ratones , Microambiente Tumoral
14.
Leukemia ; 34(6): 1658-1668, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31776464

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare aggressive myelodysplastic/myeloproliferative neoplasm of early childhood, initiated by RAS-activating mutations. Genomic analyses have recently described JMML mutational landscape; however, the nature of JMML-propagating cells (JMML-PCs) and the clonal architecture of the disease remained until now elusive. Combining genomic (exome, RNA-seq), Colony forming assay and xenograft studies, we detect the presence of JMML-PCs that faithfully reproduce JMML features including the complex/nonlinear organization of dominant/minor clones, both at diagnosis and relapse. Further integrated analysis also reveals that although the mutations are acquired in hematopoietic stem cells, JMML-PCs are not always restricted to this compartment, highlighting the heterogeneity of the disease during the initiation steps. We show that the hematopoietic stem/progenitor cell phenotype is globally maintained in JMML despite overexpression of CD90/THY-1 in a subset of patients. This study shed new lights into the ontogeny of JMML, and the identity of JMML-PCs, and provides robust models to monitor the disease and test novel therapeutic approaches.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mielomonocítica Juvenil/patología , Células Madre Neoplásicas/patología , Adolescente , Animales , Niño , Preescolar , Femenino , Xenoinjertos , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/genética , Masculino , Ratones , Mutación
15.
Nat Commun ; 11(1): 6372, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33311516

RESUMEN

The thymus is a primary lymphoid organ, essential for T cell maturation and selection. There has been long-standing interest in processes underpinning thymus generation and the potential to manipulate it clinically, because alterations of thymus development or function can result in severe immunodeficiency and autoimmunity. Here, we identify epithelial-mesenchymal hybrid cells, capable of long-term expansion in vitro, and able to reconstitute an anatomic phenocopy of the native thymus, when combined with thymic interstitial cells and a natural decellularised extracellular matrix (ECM) obtained by whole thymus perfusion. This anatomical human thymus reconstruction is functional, as judged by its capacity to support mature T cell development in vivo after transplantation into humanised immunodeficient mice. These findings establish a basis for dissecting the cellular and molecular crosstalk between stroma, ECM and thymocytes, and offer practical prospects for treating congenital and acquired immunological diseases.


Asunto(s)
Células del Estroma , Timo/inmunología , Animales , Autoinmunidad , Diferenciación Celular , Células Epiteliales/inmunología , Matriz Extracelular , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Ratas , Regeneración , Timocitos , Timo/patología , Timo/trasplante , Andamios del Tejido
16.
Dev Cell ; 4(1): 67-82, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12530964

RESUMEN

Lateral inhibition, mediated by Notch signaling, leads to the selection of cells that are permitted to become neurons within domains defined by proneural gene expression. Reduced lateral inhibition in zebrafish mib mutant embryos permits too many neural progenitors to differentiate as neurons. Positional cloning of mib revealed that it is a gene in the Notch pathway that encodes a RING ubiquitin ligase. Mib interacts with the intracellular domain of Delta to promote its ubiquitylation and internalization. Cell transplantation studies suggest that mib function is essential in the signaling cell for efficient activation of Notch in neighboring cells. These observations support a model for Notch activation where the Delta-Notch interaction is followed by endocytosis of Delta and transendocytosis of the Notch extracellular domain by the signaling cell. This facilitates intramembranous cleavage of the remaining Notch receptor, release of the Notch intracellular fragment, and activation of target genes in neighboring cells.


Asunto(s)
Ligasas/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas , Ubiquitina/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Western Blotting , Diferenciación Celular , Endocitosis , Regulación de la Expresión Génica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Ligasas/química , Ligasas/genética , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Neuronas/citología , Fenotipo , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Notch , Médula Espinal/embriología , Médula Espinal/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
17.
Mol Cell Biol ; 22(20): 7313-24, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12242306

RESUMEN

Chromosomal translocation t(9;11)(p22;q23) in acute myeloid leukemia fuses the MLL and AF9 genes. We have inactivated the murine homologue of AF9 to elucidate its normal role. No effect on hematopoiesis was observed in mice with a null mutation of Af9. However, an Af9 null mutation caused perinatal lethality, and homozygous mice exhibited anomalies of the axial skeleton. Both the cervical and thoracic regions were affected by anterior homeotic transformation. Strikingly, mice lacking functional Af9 exhibited a grossly deformed atlas and an extra cervical vertebra. To determine the molecular mediators of this phenotype, analysis of Hox gene expression by in situ hybridization showed that Af9 null embryos have posterior changes in Hoxd4 gene expression. We conclude that the Af9 gene is required for normal embryogenesis in mice by controlling pattern formation, apparently via control of Hox gene regulation. This is analogous to the role of Mll, the murine homolog of human MLL, to which the Af9 gene fuses in acute myeloid leukemias.


Asunto(s)
Tipificación del Cuerpo , Regulación de la Expresión Génica , Proteínas Nucleares/fisiología , Proto-Oncogenes , Columna Vertebral/embriología , Factores de Transcripción/genética , Animales , Huesos/embriología , Huesos/metabolismo , Proteínas de Unión al ADN , Expresión Génica , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio , Humanos , Leucemia , Ratones , Ratones Noqueados , Mutagénesis , Proteína de la Leucemia Mieloide-Linfoide , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/genética , Columna Vertebral/metabolismo , Translocación Genética
18.
J Vis Exp ; (126)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28809828

RESUMEN

Human hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, an intricate, multifactorial network of components producing cytokines, growth factors, and extracellular matrix. The ability of HSCs to remain quiescent, self-renew or differentiate, and acquire mutations and become malignant depends upon the complex interactions they establish with different stromal components. To observe the crosstalk between human HSCs and the human BM niche in physiological and pathological conditions, we designed a protocol to ectopically model and image a humanized BM niche in immunodeficient mice. We show that the use of different cellular components allows for the formation of humanized structures and the opportunity to sustain long-term human hematopoietic engraftment. Using two-photon microscopy, we can live-image these structures in situ at the single-cell resolution, providing a powerful new tool for the functional characterization of the human BM microenvironment and its role in regulating normal and malignant hematopoiesis.


Asunto(s)
Bioingeniería/métodos , Médula Ósea/fisiología , Células Madre Hematopoyéticas/fisiología , Imagen Óptica/métodos , Andamios del Tejido , Animales , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Colágeno/química , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones SCID , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Imagen Óptica/instrumentación , Nicho de Células Madre/fisiología
19.
Cancer Cell ; 32(3): 324-341.e6, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28870739

RESUMEN

The biological and clinical behaviors of hematological malignancies can be influenced by the active crosstalk with an altered bone marrow (BM) microenvironment. In the present study, we provide a detailed picture of the BM vasculature in acute myeloid leukemia using intravital two-photon microscopy. We found several abnormalities in the vascular architecture and function in patient-derived xenografts (PDX), such as vascular leakiness and increased hypoxia. Transcriptomic analysis in endothelial cells identified nitric oxide (NO) as major mediator of this phenotype in PDX and in patient-derived biopsies. Moreover, induction chemotherapy failing to restore normal vasculature was associated with a poor prognosis. Inhibition of NO production reduced vascular permeability, preserved normal hematopoietic stem cell function, and improved treatment response in PDX.


Asunto(s)
Antineoplásicos/uso terapéutico , Médula Ósea/patología , Permeabilidad Capilar , Microambiente Celular , Progresión de la Enfermedad , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Trasplante de Neoplasias/patología , Óxido Nítrico/metabolismo , Resultado del Tratamiento
20.
Front Cell Neurosci ; 9: 74, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25814931

RESUMEN

The Notch signaling pathway controls differentiation of hair cells and supporting cells in the vertebrate inner ear. Here, we have investigated whether Numb, a known regulator of Notch activity in Drosophila, is involved in this process in the embryonic chick. The chicken homolog of Numb is expressed throughout the otocyst at early stages of development and is concentrated at the basal pole of the cells. It is asymmetrically allocated at some cell divisions, as in Drosophila, suggesting that it could act as a determinant inherited by one of the two daughter cells and favoring adoption of a hair-cell fate. To test the implication of Numb in hair cell fate decisions and the regulation of Notch signaling, we used different methods to overexpress Numb at different stages of inner ear development. We found that sustained or late Numb overexpression does not promote hair cell differentiation, and Numb does not prevent the reception of Notch signaling. Surprisingly, none of the Numb-overexpressing cells differentiated into hair cells, suggesting that high levels of Numb protein could interfere with intracellular processes essential for hair cell survival. However, when Numb was overexpressed early and more transiently during ear development, no effect on hair cell formation was seen. These results suggest that in the inner ear at least, Numb does not significantly repress Notch activity and that its asymmetric distribution in dividing precursor cells does not govern the choice between hair cell and supporting cell fates.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA