Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Leukemia ; 36(11): 2634-2646, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36163264

RESUMEN

Disease progression and relapse of chronic myeloid leukemia (CML) are caused by therapy resistant leukemia stem cells (LSCs), and cure relies on their eradication. The microenvironment in the bone marrow (BM) is known to contribute to LSC maintenance and resistance. Although leukemic infiltration of the spleen is a hallmark of CML, it is unknown whether spleen cells form a niche that maintains LSCs. Here, we demonstrate that LSCs preferentially accumulate in the spleen and contribute to disease progression. Spleen LSCs were located in the red pulp close to red pulp macrophages (RPM) in CML patients and in a murine CML model. Pharmacologic and genetic depletion of RPM reduced LSCs and decreased their cell cycling activity in the spleen. Gene expression analysis revealed enriched stemness and decreased myeloid lineage differentiation in spleen leukemic stem and progenitor cells (LSPCs). These results demonstrate that splenic RPM form a niche that maintains CML LSCs in a quiescent state, resulting in disease progression and resistance to therapy.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Leucemia Mieloide , Humanos , Ratones , Animales , Bazo , Células Madre Neoplásicas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mieloide/genética , Macrófagos/metabolismo , Progresión de la Enfermedad , Microambiente Tumoral
2.
Nat Mach Intell ; 3(9): 799-811, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34541455

RESUMEN

Fluorescence microscopy allows for a detailed inspection of cells, cellular networks, and anatomical landmarks by staining with a variety of carefully-selected markers visualized as color channels. Quantitative characterization of structures in acquired images often relies on automatic image analysis methods. Despite the success of deep learning methods in other vision applications, their potential for fluorescence image analysis remains underexploited. One reason lies in the considerable workload required to train accurate models, which are normally specific for a given combination of markers, and therefore applicable to a very restricted number of experimental settings. We herein propose Marker Sampling and Excite - a neural network approach with a modality sampling strategy and a novel attention module that together enable (i) flexible training with heterogeneous datasets with combinations of markers and (ii) successful utility of learned models on arbitrary subsets of markers prospectively. We show that our single neural network solution performs comparably to an upper bound scenario where an ensemble of many networks is naïvely trained for each possible marker combination separately. In addition, we demonstrate the feasibility of this framework in high-throughput biological analysis by revising a recent quantitative characterization of bone marrow vasculature in 3D confocal microscopy datasets and further confirm the validity of our approach on an additional, significantly different dataset of microvessels in fetal liver tissues. Not only can our work substantially ameliorate the use of deep learning in fluorescence microscopy analysis, but it can also be utilized in other fields with incomplete data acquisitions and missing modalities.

3.
J Exp Med ; 218(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34709350

RESUMEN

Chronic viral infections are associated with hematopoietic suppression, bone marrow (BM) failure, and hematopoietic stem cell (HSC) exhaustion. However, how persistent viral challenge and inflammatory responses target BM tissues and perturb hematopoietic competence remains poorly understood. Here, we combine functional analyses with advanced 3D microscopy to demonstrate that chronic infection with lymphocytic choriomeningitis virus leads to (1) long-lasting decimation of the BM stromal network of mesenchymal CXCL12-abundant reticular cells, (2) proinflammatory transcriptional remodeling of remaining components of this key niche subset, and (3) durable functional defects and decreased competitive fitness in HSCs. Mechanistically, BM immunopathology is elicited by virus-specific, activated CD8 T cells, which accumulate in the BM via interferon-dependent mechanisms. Combined antibody-mediated inhibition of type I and II IFN pathways completely preempts degeneration of CARc and protects HSCs from chronic dysfunction. Hence, viral infections and ensuing immune reactions durably impact BM homeostasis by persistently decreasing the competitive fitness of HSCs and disrupting essential stromal-derived, hematopoietic-supporting cues.


Asunto(s)
Médula Ósea/virología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/virología , Coriomeningitis Linfocítica/patología , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Enfermedad Crónica , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Interferones/metabolismo , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Ratones Endogámicos C57BL , Ratones Mutantes , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo
4.
Ann N Y Acad Sci ; 1466(1): 5-16, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31368140

RESUMEN

Hematopoietic stem cells (HSCs) have been long proposed to reside in defined anatomical locations within bone marrow (BM) tissues in direct contact or close proximity to nurturing cell types. Imaging techniques that allow the simultaneous mapping of HSCs and interacting cell types have been central to the discovery of basic principles of these so-called HSC niches. Despite major progress in the field, a quantitative and comprehensive model of the cellular and molecular components that define these specialized microenvironments is lacking to date, and uncertainties remain on the preferential localization of HSCs in the context of complex BM tissue landscapes. Recent technological breakthroughs currently allow for the quantitative spatial analysis of BM cellular components with extraordinary precision. Here, we critically discuss essential technical aspects related to imaging approaches, image processing tools, and spatial statistics, which constitute the three basic elements of rigorous quantitative spatial analyses of HSC niches in the BM microenvironment.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Imagen Molecular/métodos , Nicho de Células Madre/fisiología , Animales , Médula Ósea/diagnóstico por imagen , Médula Ósea/fisiología , Microambiente Celular/fisiología , Diagnóstico por Imagen/métodos , Humanos , Procesamiento de Imagen Asistido por Computador/tendencias , Invenciones/tendencias , Imagen Molecular/tendencias , Análisis Espacial
5.
Cell Stem Cell ; 24(5): 769-784.e6, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30905620

RESUMEN

Chronic myeloid leukemia (CML) originates in a hematopoietic stem cell (HSC) transformed by the breakpoint cluster region (BCR)-abelson (ABL) oncogene and is effectively treated with tyrosine kinase inhibitors (TKIs). TKIs do not eliminate disease-propagating leukemic stem cells (LSCs), suggesting a deeper understanding of niche-dependent regulation of CML LSCs is required to eradicate disease. Cxcl12 is expressed in bone marrow niches and controls HSC maintenance, and here, we show that targeted deletion of Cxcl12 from mesenchymal stromal cells (MSCs) reduces normal HSC numbers but promotes LSC expansion by increasing self-renewing cell divisions, possibly through enhanced Ezh2 activity. In contrast, endothelial cell-specific Cxcl12 deletion decreases LSC proliferation, suggesting niche-specific effects. During CML development, abnormal clusters of colocalized MSCs and LSCs form but disappear upon Cxcl12 deletion. Moreover, MSC-specific deletion of Cxcl12 increases LSC elimination by TKI treatment. These findings highlight a critical role of niche-specific effects of Cxcl12 expression in maintaining quiescence of TKI-resistant LSC populations.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Células Madre Mesenquimatosas/fisiología , Células Madre Neoplásicas/fisiología , Nicho de Células Madre/fisiología , Animales , Apoptosis , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Resistencia a Antineoplásicos , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Inhibidores de Proteínas Quinasas/uso terapéutico
6.
Nat Commun ; 9(1): 2532, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29955044

RESUMEN

Sinusoidal endothelial cells and mesenchymal CXCL12-abundant reticular cells are principal bone marrow stromal components, which critically modulate haematopoiesis at various levels, including haematopoietic stem cell maintenance. These stromal subsets are thought to be scarce and function via highly specific interactions in anatomically confined niches. Yet, knowledge on their abundance, global distribution and spatial associations remains limited. Using three-dimensional quantitative microscopy we show that sinusoidal endothelial and mesenchymal reticular subsets are remarkably more abundant than estimated by conventional flow cytometry. Moreover, both cell types assemble in topologically complex networks, associate to extracellular matrix and pervade marrow tissues. Through spatial statistical methods we challenge previous models and demonstrate that even in the absence of major specific interaction forces, virtually all tissue-resident cells are invariably in physical contact with, or close proximity to, mesenchymal reticular and sinusoidal endothelial cells. We further show that basic structural features of these stromal components are preserved during ageing.


Asunto(s)
Envejecimiento/fisiología , Células de la Médula Ósea/ultraestructura , Fémur/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/ultraestructura , Células Madre Mesenquimatosas/ultraestructura , Animales , Médula Ósea/diagnóstico por imagen , Médula Ósea/fisiología , Células de la Médula Ósea/fisiología , Recuento de Células , Movimiento Celular , Microambiente Celular/fisiología , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Fémur/diagnóstico por imagen , Fémur/fisiología , Células Madre Hematopoyéticas/fisiología , Imagenología Tridimensional/estadística & datos numéricos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Microscopía/métodos , Nicho de Células Madre
7.
Front Immunol ; 7: 689, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28163704

RESUMEN

Continuous production of blood cells unfolds within a complex three-dimensional tissue scaffold established by highly organized stromal cell networks of mesenchymal, neural, and vascular origin inside bone marrow (BM) cavities. Collectively, stromal cells have been shown to serve two principal roles; first as primary participants of bone remodeling and metabolism and second as master regulators of different stages of blood cell development and production. Indeed, ample evidence demonstrates that stromal cells can sense and integrate systemic signals to shape hematopoietic responses and that these regulatory mechanisms are subverted in multiple pathologic conditions. Microbial infections are stressors that elicit potent inflammatory reactions and induce substantial alterations of hematopoietic output. Whether the cellular components of the BM stromal microenvironment are targeted by infections and participate in infection-induced hematopoiesis has not been investigated in sufficient detail to date. In this manuscript, we provide a succinct updated overview of the different cell populations that are currently known to form BM stroma. We discuss experimental evidence demonstrating that different stromal components are actively damaged or functionally altered by pathogens and/or ensuing inflammatory signals and review how these effects are known to contribute to the hematologic manifestations observed during infections.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA