Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38617223

RESUMEN

Hematopoietic dysfunction has been associated with a reduction in the number of active precursors. However, precursor quantification at homeostasis and under diseased conditions is constrained by the scarcity of available methods. To address this issue, we optimized a method for quantifying a wide range of hematopoietic precursors. Assuming the random induction of a stable label in precursors following a binomial distribution, the estimation depends on the inverse correlation between precursor numbers and the variance of precursor labeling among independent samples. Experimentally validated to cover the full dynamic range of hematopoietic precursors in mice (1 to 105), we utilized this approach to demonstrate that thousands of precursors, which emerge after modest expansion during fetal-to-adult transition, contribute to native and perturbed hematopoiesis. We further estimated the number of precursors in a mouse model of Fanconi Anemia, showcasing how repopulation deficits can be segregated into autologous (cell proliferation) and non-autologous causes (lack of precursor). Our results support an accessible and reliable approach for precursor quantification, emphasizing the contemporary perspective that native hematopoiesis is highly polyclonal.

2.
3.
Haematologica ; 2023 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-37855027

RESUMEN

A dysfunctional immune tumour microenvironment facilitates disease progression in multiple myeloma (MM). Using multiplex immunohistochemistry (mIHC), we described the quantitative and qualitative changes in CD3+ CD8+ cytotoxic T-cells and assess their proximity to malignant plasma cells (PCs) in patients with monoclonal gammopathy of undetermined significance (MGUS), newly diagnosed (ND) and relapsed/refractory (RR) MM. Formalin-fixed, paraffin-embedded trephine sections from patients with MGUS (n=32), NDMM (n=65) and RRMM (n=59) were sequentially stained for CD138, CD3, CD8, and checkpoint receptors (CPR) Tim-3, Lag-3, and PD-1. Halo® image analysis platform was used for cell segmentation and phenotyping, facilitating enumeration of cytotoxic T-cells and analysis of proximity to PCs. The percentage of CD8+ cytotoxic T-cells in proximity to PCs is greater in patients with NDMM than patients with RRMM (at 50gm distance 90.8% vs. 81.5%, p=0.038). There is a trend for more CD3+ T-cells in MGUS (p=0.08) but no difference was observed in the prevalence of CD8+ cytotoxic T-cells (p=0.48). Lag-3 is the most common CPR expressed on cytotoxic T-cells in myeloma (p.

4.
Exp Hematol ; 116: 1-14, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36283572

RESUMEN

The founder of all blood cells are hematopoietic stem cells (HSCs), which are rare stem cells that undergo key cell fate decisions to self-renew to generate more HSCs or to differentiate progressively into a hierarchy of different immature hematopoietic cell types to ultimately produce mature blood cells. These decisions are influenced both intrinsically and extrinsically, the latter by microenvironment cells in the bone marrow (BM). In recent decades, notable progress in our ability to identify, isolate, and study key properties of adult murine HSCs and multipotent progenitor (MPP) cells has challenged our prior understanding of the hierarchy of these primitive hematopoietic cells. These studies have revealed the existence of at least two distinct HSC types in adults: one that generates all hematopoietic cell lineages with almost equal potency and one that is platelet/myeloid-biased and increases with aging. These studies have also revealed distinct MPP cell types that have different functional potential. This review provides an update to these murine HSCs and MPP cells, their key functional properties, and the assays that have been used to assess their potential.


Asunto(s)
Células Madre Hematopoyéticas , Células Madre Multipotentes , Ratones , Animales , Linaje de la Célula , Células Madre Hematopoyéticas/metabolismo , Células Madre Multipotentes/metabolismo , Diferenciación Celular , Médula Ósea
5.
Immunol Cell Biol ; 100(9): 674-678, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35748687

RESUMEN

Universally, women are under-represented in senior academic leadership in science, technology, engineering, maths and medicine (STEMM). Successful funding outcomes are a critical point in career progression, to continue both a scientist's research but also for their retention within the STEMM workforce. A common explanation for the lower success rate of women in securing funding is that fewer women apply for funding. However, this does not adequately explain the gender inequities in funding outcomes, both in terms of fewer funded applications and also of reduced funding awarded per grant, resulting in less overall success. Gendered funding outcomes occur within academic institutions and peak funding bodies due to historical, systemic conscious and unconscious biases during peer review. As a cumulative bias over a woman's research career, this results in women being under-represented in STEMM and the loss of their contributions to medical research, reducing innovation through a lack of diverse workforces.


Asunto(s)
Distinciones y Premios , Investigación Biomédica , Australia , Femenino , Humanos , Liderazgo , Tecnología
6.
J Bone Miner Res ; 37(7): 1321-1334, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35490308

RESUMEN

Osteoblasts and their progenitors play an important role in the support of hematopoiesis within the bone marrow (BM) microenvironment. We have previously reported that parathyroid hormone receptor (PTH1R) signaling in osteoprogenitors is required for normal B cell precursor differentiation, and for trafficking of maturing B cells out of the BM. Cells of the osteoblast lineage have been implicated in the regulation of several other hematopoietic cell populations, but the effects of PTH1R signaling in osteoprogenitors on other maturing hematopoietic populations have not been investigated. Here we report that numbers of maturing myeloid, T cell, and erythroid populations were increased in the BM of mice lacking PTH1R in Osx-expressing osteoprogenitors (PTH1R-OsxKO mice; knockout [KO]). This increase in maturing hematopoietic populations was not associated with an increase in progenitor populations or proliferation. The spleens of PTH1R-OsxKO mice were small with decreased numbers of all hematopoietic populations, suggesting that trafficking of mature hematopoietic populations between BM and spleen is impaired in the absence of PTH1R in osteoprogenitors. RNA sequencing (RNAseq) of osteoprogenitors and their descendants in bone and BM revealed increased expression of vascular cell adhesion protein 1 (VCAM-1) and C-X-C motif chemokine ligand 12 (CXCL12), factors that are involved in trafficking of several hematopoietic populations. © 2022 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Osteoblastos , Receptor de Hormona Paratiroídea Tipo 1 , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Osteoblastos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transducción de Señal
7.
JBMR Plus ; 5(10): e10516, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34693187

RESUMEN

The bone marrow microenvironment (BMME) regulates hematopoiesis through a complex network of cellular and molecular components. Hematologic malignancies reside within, and extensively interact with, the same BMME. These interactions consequently alter both malignant and benign hematopoiesis in multiple ways, and can encompass initiation of malignancy, support of malignant progression, resistance to chemotherapy, and loss of normal hematopoiesis. Herein, we will review supporting studies for interactions of the BMME with hematologic malignancies and discuss challenges still facing this exciting field of research. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

9.
Exp Hematol ; 100: 41-51, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34228982

RESUMEN

Bone marrow stromal cells (BMSCs) are a key part of the hematopoietic niche. Mouse and human BMSCs are recognized by different markers (LepR and NGFR/CD271, respectively). However, there has not been a detailed in situ comparison of both populations within the hematopoietic microenvironment. Moreover, dog BMSCs have not been characterized in situ by any of those markers. We conducted a systematic histopathological comparison of mouse, human, and dog BMSCs within their bone marrow architecture and microenvironment. Human and dog CD271+ BMSCs had a morphology, frequency, and distribution within trabecular bone marrow similar to those of mouse LepR+ BMSCs. However, mouse bone marrow had higher cellularity and megakaryocyte content. In conclusion, highly comparable bone marrow mesenchymal stromal cell distribution among the three species establishes the validity of using mouse and dog as a surrogate experimental model of hematopoietic stem cell-BMSC interactions. However, the distinct differences in adipocyte and megakaryocyte microenvironment content of mouse bone marrow and how they might influence hematopoietic stem cell interactions as compared with humans require further study.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Células Cultivadas , Niño , Perros , Humanos , Células Madre Mesenquimatosas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteínas del Tejido Nervioso/análisis , Receptores de Leptina/análisis , Receptores de Factor de Crecimiento Nervioso/análisis , Especificidad de la Especie , Nicho de Células Madre
10.
Immunol Rev ; 302(1): 47-67, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34002391

RESUMEN

B lymphocytes are crucial for the body's humoral immune response, secreting antibodies generated against foreign antigens to fight infection. Adult murine B lymphopoiesis is initiated in the bone marrow and additional maturation occurs in the spleen. In both these organs, B lymphopoiesis involves interactions with numerous different non-hematopoietic cells, also known as stromal or microenvironment cells, which provide migratory, maturation, and survival signals. A variety of conditional knockout and transgenic mouse models have been used to identify the roles of distinct microenvironment cell types in the regulation of B lymphopoiesis. These studies have revealed that mesenchymal lineage cells and endothelial cells comprise the non-hematopoietic microenvironment cell types that support B lymphopoiesis in the bone marrow. In the spleen, various types of stromal cells and endothelial cells contribute to B lymphocyte maturation. More recently, comprehensive single cell RNA-seq studies have also been used to identify clusters of stromal cell types in the bone marrow and spleen, which will aid in further identifying key regulators of B lymphopoiesis. Here, we review the different types of microenvironment cells and key extrinsic regulators that are known to be involved in the regulation of murine B lymphopoiesis in the bone marrow and spleen.


Asunto(s)
Células Endoteliales , Linfopoyesis , Animales , Linfocitos B , Médula Ósea , Células de la Médula Ósea , Ratones , Células del Estroma
11.
Blood ; 138(4): 304-317, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-33786586

RESUMEN

Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited because of a lack of methods to isolate these cells. We found that murine Lin-CD31-Sca-1-CD51+ cells can be divided into 4 subpopulations by using flow cytometry based on their expression of the platelet-derived growth factor receptors ⍺ and ß (PDGFR⍺ and PDGFRß). The use of different skeletal lineage reporters confirmed the skeletal origin of the 4 populations. Multiplex immunohistochemistry studies revealed that all 4 populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit-fibroblast capacity, and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the 4 populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that 1 of these 4 different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B-lymphocyte precursors were also observed in these distinct skeletal sites. This cell population also supported pre-B lymphopoiesis in culture. Our method of isolating 4 distinct maturing skeletal populations will help elucidate the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linfopoyesis/inmunología , Músculo Esquelético/inmunología , Animales , Diferenciación Celular/genética , Linfopoyesis/genética , Ratones , Ratones Transgénicos
12.
Dev Cell ; 55(2): 109-111, 2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33108749

RESUMEN

The generation of adult hematopoietic stem and progenitor cells (HSPCs) from embryonic and induced pluripotent stem cells (iPSCs) will provide therapeutic benefits but is currently elusive. In this issue of Developmental Cell, Frame et al. reveal that the inflammasome has a key role in HSPCs specification during endothelial-to-hematopoietic transition (EHT).


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inflamasomas , Células Sanguíneas , Diferenciación Celular , Hematopoyesis , Azúcares
13.
Cell Cycle ; 19(20): 2573-2588, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32900260

RESUMEN

Acute myeloid leukemia (AML) is an aggressive, often fatal hematopoietic malignancy. All-trans retinoic acid (atRA), one of the first molecularly targeted drugs in oncology, has greatly improved the outcome of a subtype of AML, acute promyelocytic leukemia (APL). In contrast, atRA has so far provided little therapeutic benefit in the much larger group of patients with non-APL AML. Attempts to identify genetically or molecularly defined subgroups of patients that may respond to atRA have not yielded consistent results. Since AML is a stem cell-driven disease, understanding the effectiveness of atRA may require an appreciation of its impact on AML stem cells. Recent studies reported that atRA decreased stemness of AML with an FLT3-ITD mutation, yet increased it in AML1-ETO driven or EVI1-overexpressing AML. This review summarizes the role of atRA in normal hematopoiesis and in AML, focusing on its impact on AML stem cells.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Tretinoina/farmacología , Animales , Apoptosis/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Humanos
14.
Exp Hematol ; 82: 1-7, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32006607

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Therapies for pediatric ALL have improved such that more than 80% of patients survive to 5 years post-therapy, and most survive to adulthood. These ALL patients experience long-term side effects that permanently affect their quality of life, with bone loss and reduced longitudinal growth being the most common skeletal complications. To determine the effects of the chemotherapeutic agents used in ALL induction therapy on bone density and longitudinal growth in mice, we treated juvenile mice with doxorubicin, dexamethasone, vincristine, l-asparaginase, or combination therapy. At adulthood, mice were culled and bones collected and scanned by micro-computed tomography (micro-CT). Mice that received doxorubicin and combination therapy exhibited reduced longitudinal growth and significant reductions in trabecular bone volume, trabecular thickness, and trabecular number, with increased trabecular separation. Mean cortical thickness, cortical area, marrow area, endocortical perimeter, and polar moment of inertia were significantly reduced by doxorubicin and combination therapy. Vincristine treatment significantly decreased trabecular bone volume, trabecular number, and increased trabecular separation but had no effects on cortical bone. Dexamethasone treatment increased trabecular bone separation, cortical marrow area, and cortical bone periosteal perimeter. Mice treated with l-asparaginase did not have any bone phenotypes. In conclusion, these data indicate that the majority of the chemotherapy agents used in induction therapy for pediatric ALL have long-term effects on bone in mice. A single dose of doxorubicin in juvenile mice was sufficient to cause the majority of the bone phenotypes, with combination therapy intensifying these effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Hueso Esponjoso , Placa de Crecimiento , Quimioterapia de Inducción/efectos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Microtomografía por Rayos X , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Asparaginasa/efectos adversos , Asparaginasa/farmacología , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/crecimiento & desarrollo , Niño , Dexametasona/efectos adversos , Dexametasona/farmacología , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Placa de Crecimiento/diagnóstico por imagen , Placa de Crecimiento/crecimiento & desarrollo , Humanos , Masculino , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico por imagen , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Vincristina/efectos adversos , Vincristina/farmacología
15.
Cell Death Dis ; 10(12): 944, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822659

RESUMEN

Ecotropic virus integration site 1 (EVI1), whose overexpression characterizes a particularly aggressive subtype of acute myeloid leukemia (AML), enhanced anti-leukemic activities of all-trans retinoic acid (atRA) in cell lines and patient samples. However, the drivers of leukemia formation, therapy resistance, and relapse are leukemic stem cells (LSCs), whose properties were hardly reflected in these experimental setups. The present study was designed to address the effects of, and interactions between, EVI1 and retinoids in AML LSCs. We report that Evi1 reduced the maturation of leukemic cells and promoted the abundance, quiescence, and activity of LSCs in an MLL-AF9-driven mouse model of AML. atRA further augmented these effects in an Evi1 dependent manner. EVI1 also strongly enhanced atRA regulated gene transcription in LSC enriched cells. One of their jointly regulated targets, Notch4, was an important mediator of their effects on leukemic stemness. In vitro exposure of leukemic cells to a pan-RAR antagonist caused effects opposite to those of atRA. In vivo antagonist treatment delayed leukemogenesis and reduced LSC abundance, quiescence, and activity in Evi1high AML. Key results were confirmed in human myeloid cell lines retaining some stem cell characteristics as well as in primary human AML samples. In summary, our study is the first to report the importance of EVI1 for key properties of AML LSCs. Furthermore, it shows that atRA enhances, and a pan-RAR antagonist counteracts, the effects of EVI1 on AML stemness, thus raising the possibility of using RAR antagonists in the therapy of EVI1high AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Receptor Notch4/genética , Tretinoina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Células Mieloides/efectos de los fármacos , Células Madre Neoplásicas/metabolismo
16.
Cell Tissue Res ; 378(3): 441-456, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31302761

RESUMEN

Artemin is a neurotrophic factor that plays a crucial role in the regulation of neural development and regeneration and has also been implicated in the pathogenesis of inflammatory pain. The receptor for artemin, GFRα3, is expressed by sympathetic and nociceptive sensory neurons, including some that innervate the bone marrow, but it is unclear if it is also expressed in other cell types in the bone marrow. Our goal in the present study was to characterise the expression of GFRα3 in nonneuronal cells in the bone marrow. Immunohistochemical studies revealed that GFRα3-expressing cells in the bone marrow are spatially associated with blood vessels and are in intimate contact with nerve fibres. We used various combinations of markers to distinguish different cell types and found that the GFRα3-expressing cells expressed markers of nonmyelinating Schwann cells (e.g. GFAP, p75NTR, nestin). Analysis of bone marrow sections of Wnt1-reporter mice also demonstrated that they originate from the neural crest. Further characterisation using flow cytometry revealed that GFRα3 is expressed in a population of CD51+Sca1-PDGFRα- cells, reinforcing the notion that they are neural crest-derived, nonmyelinating Schwann cells. In conclusion, there is a close association between peripheral nerve terminals and a population of nonneuronal cells that express GFRα3 in the bone marrow. The nonneuronal cells have characteristics consistent with a neural crest-derived, nonmyelinating Schwann cell phenotype. Our findings provide a better understanding of the expression pattern of GFRα3 in the bone marrow microenvironment.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células de Schwann/metabolismo , Animales , Células de la Médula Ósea/citología , Ratones , Ratones Endogámicos C57BL , Células de Schwann/citología
17.
Sci Rep ; 9(1): 7050, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31065022

RESUMEN

Deletion of long arm of chromosome 20 [del(20q)] is the second most frequent recurrent chromosomal abnormality in hematological malignancies. It is detected in 10% of myeloproliferative neoplasms, 4-5% of myelodysplastic syndromes, and 1-2% of acute myeloid leukaemia. Recurrent, non-random occurrence of del(20q) indicates that it is a pathogenic driver in myeloid malignancies. Genetic mapping of patient samples has identified two regions of interest on 20q - the "Common Deleted Region" (CDR) and "Common Retained Region" (CRR), which was often amplified. We proposed that the CDR contained tumor suppressor gene(s) (TSG) and the CRR harbored oncogene(s); loss of a TSG together with over-expression of an oncogene favored development of myeloid malignancies. Protein Tyrosine Phosphatase Receptor T (PTPRT) and Hemopoietic cell kinase (HCK) were identified to be the likely candidate TSG and oncogene respectively. Retroviral transduction of HCK into PTPRT-null murine LKS+ stem and progenitor cells resulted in hyperproliferation in colony forming assays and hyperphosphorylation of intracellular STAT3. Furthermore, over half of the murine recipients of these transduced cells developed erythroid hyperplasia, polycythemia and splenomegaly at 12 months, although no leukemic phenotype was observed. The findings suggested that HCK amplification coupled with PTPRT loss in del(20q) leads to development of a myeloproliferative phenotype.


Asunto(s)
Eritropoyesis/fisiología , Policitemia/genética , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Esplenomegalia/etiología , Animales , Médula Ósea/patología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Ratones Endogámicos C57BL , Ratones Mutantes , Oncogenes , Proteínas Proto-Oncogénicas c-hck/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Factor de Transcripción STAT3/metabolismo , Esplenomegalia/patología
18.
Bone ; 119: 42-56, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29183783

RESUMEN

Early B lymphopoiesis occurs in the bone marrow and is reliant on interactions with numerous cell types in the bone marrow microenvironment, particularly those of the mesenchymal lineage. Each cellular niche that supports the distinct stages of B lymphopoiesis is unique. Different cell types and signaling molecules are important for the progressive stages of B lymphocyte differentiation. Cells expressing CXCL12 and IL-7 have long been recognized as having essential roles in facilitating progression through stages of B lymphopoiesis. Recently, a number of other factors that extrinsically mediate B lymphopoiesis (positively or negatively) have been identified. In addition, the use of transgenic mouse models to delete specific genes in mesenchymal lineage cells has further contributed to our understanding of how B lymphopoiesis is regulated in the bone marrow. This review will cover the current understanding of B lymphocyte niches in the bone marrow and key extrinsic molecules and signaling pathways involved in these niches, with a focus on how mesenchymal lineage cells regulate B lymphopoiesis.


Asunto(s)
Linfocitos B/citología , Linaje de la Célula , Células Madre Mesenquimatosas/citología , Animales , Médula Ósea/fisiología , Microambiente Celular , Humanos , Linfopoyesis , Células Madre Mesenquimatosas/metabolismo
19.
Bone ; 119: 19-35, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29704697

RESUMEN

Bone marrow contains numerous different cell types arising from hematopoietic stem cells (HSCs) and non-hematopoietic mesenchymal/skeletal stem cells, in addition to other cell types such as endothelial cells- these non-hematopoietic cells are commonly referred to as stromal cells or microenvironment cells. HSC function is intimately linked to complex signals integrated by their niches, formed by combinations of hematopoietic and stromal cells. Studies of hematopoietic cells have been significantly advanced by flow cytometry methods, enabling the quantitation of each cell type in normal and perturbed situations, in addition to the isolation of these cells for molecular and functional studies. Less is known, however, about the specific niches for distinct developing hematopoietic lineages, or the changes occurring in the niche size and function in these distinct anatomical sites in the bone marrow under stress situations and ageing. Significant advances in imaging technology during the last decade have permitted studies of HSC niches in mice. Additional imaging technologies are emerging that will facilitate the study of human HSC niches in trephine BM biopsies. Here we provide an overview of imaging technologies used to study HSC niches, in addition to highlighting emerging technology that will help us to more precisely identify and characterize HSC niches in normal and diseased states.


Asunto(s)
Células Madre Hematopoyéticas/citología , Imagen Molecular/métodos , Nicho de Células Madre , Animales , Médula Ósea/fisiología , Humanos , Imagenología Tridimensional , Ratones , Análisis de Matrices Tisulares
20.
Exp Hematol ; 70: 10-23, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30408513

RESUMEN

Myelodysplastic syndromes (MDS) and related myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) are clonal stem cell disorders, primarily affecting patients over 65 years of age. Mapping of the MDS and MDS/MPN genome identified recurrent heterozygous mutations in the RNA splicing machinery, with the SF3B1, SRSF2, and U2AF1 genes being frequently mutated. To better understand how spliceosomal mutations contribute to MDS pathogenesis in vivo, numerous groups have sought to establish conditional murine models of SF3B1, SRSF2, and U2AF1 mutations. The high degree of conservation of hematopoiesis between mice and human and the well-established phenotyping and genetic modification approaches make murine models an effective tool with which to study how a gene mutation contributes to disease pathogenesis. The murine models of spliceosomal mutations described to date recapitulate human MDS or MDS/MPN to varying extents. Reasons for the differences in phenotypes reported between alleles of the same mutation are varied, but the nature of the genetic modification itself and subsequent analysis methods are important to consider. In this review, we summarize recently reported murine models of SF3B1, SRSF2, and U2AF1 mutations, with a particular focus on the genetically engineered modifications underlying the models and the experimental approaches applied.


Asunto(s)
Neoplasias Hematológicas , Hematopoyesis/genética , Mutación , Síndromes Mielodisplásicos , Neoplasias Experimentales , Factores de Empalme de ARN , Empalmosomas , Animales , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Ratones , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Empalmosomas/genética , Empalmosomas/metabolismo , Empalmosomas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...