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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
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.
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
5.
Nature ; 538(7626): 518-522, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27750279

RESUMEN

It is widely accepted that complex interactions between cancer cells and their surrounding microenvironment contribute to disease development, chemo-resistance and disease relapse. In light of this observed interdependency, novel therapeutic interventions that target specific cancer stroma cell lineages and their interactions are being sought. Here we studied a mouse model of human T-cell acute lymphoblastic leukaemia (T-ALL) and used intravital microscopy to monitor the progression of disease within the bone marrow at both the tissue-wide and single-cell level over time, from bone marrow seeding to development/selection of chemo-resistance. We observed highly dynamic cellular interactions and promiscuous distribution of leukaemia cells that migrated across the bone marrow, without showing any preferential association with bone marrow sub-compartments. Unexpectedly, this behaviour was maintained throughout disease development, from the earliest bone marrow seeding to response and resistance to chemotherapy. Our results reveal that T-ALL cells do not depend on specific bone marrow microenvironments for propagation of disease, nor for the selection of chemo-resistant clones, suggesting that a stochastic mechanism underlies these processes. Yet, although T-ALL infiltration and progression are independent of the stroma, accumulated disease burden leads to rapid, selective remodelling of the endosteal space, resulting in a complete loss of mature osteoblastic cells while perivascular cells are maintained. This outcome leads to a shift in the balance of endogenous bone marrow stroma, towards a composition associated with less efficient haematopoietic stem cell function. This novel, dynamic analysis of T-ALL interactions with the bone marrow microenvironment in vivo, supported by evidence from human T-ALL samples, highlights that future therapeutic interventions should target the migration and promiscuous interactions of cancer cells with the surrounding microenvironment, rather than specific bone marrow stroma, to combat the invasion by and survival of chemo-resistant T-ALL cells.


Asunto(s)
Células de la Médula Ósea/citología , Leucemia-Linfoma de Células T del Adulto/patología , Trasplante de Neoplasias , Microambiente Tumoral , Animales , Movimiento Celular , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Microscopía Intravital , Masculino , Ratones , Osteoblastos/citología , Análisis de la Célula Individual
6.
Blood ; 132(6): 608-621, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903888

RESUMEN

Mutations in SRSF2 occur in myelodysplastic syndromes (MDS) and MDS/myeloproliferative neoplasms (MPN). SRSF2 mutations cluster at proline 95, with the most frequent mutation being a histidine (P95H) substitution. They undergo positive selection, arise early in the course of disease, and have been identified in age-related clonal hemopoiesis. It is not clear how mutation of SRSF2 modifies hemopoiesis or contributes to the development of myeloid bias or MDS/MPN. Two prior mouse models of Srsf2P95H mutation have been reported; however, these models do not recapitulate many of the clinical features of SRSF2-mutant disease and relied on bone marrow (BM) transplantation stress to elicit the reported phenotypes. We describe a new conditional murine Srsf2P95H mutation model, where the P95H mutation is expressed physiologically and heterozygously from its endogenous locus after Cre activation. Using multiple Cre lines, we demonstrate that during native hemopoiesis (ie, no BM transplantation), the Srsf2P95H mutation needs to occur within the hemopoietic stem-cell-containing populations to promote myelomonocytic bias and expansion with corresponding transcriptional and RNA splicing changes. With age, nontransplanted Srsf2P95H animals developed a progressive, transplantable disease characterized by myeloid bias, morphological dysplasia, and monocytosis, hallmarks of MDS/MPN in humans. Analysis of cooccurring mutations within the BM demonstrated the acquisition of additional mutations that are recurrent in humans with SRSF2 mutations. The tractable Srsf2P95H/+ knock-in model we have generated is highly relevant to human disease and will serve to elucidate the effect of SRSF2 mutations on initiation and maintenance of MDS/MPN.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Síndromes Mielodisplásicos/genética , Células Mieloides/metabolismo , Mielopoyesis/genética , Trastornos Mieloproliferativos/genética , Factores de Empalme Serina-Arginina/genética , Envejecimiento/genética , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Exoma , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes p53 , Células Madre Hematopoyéticas/patología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Empalme del ARN , Quimera por Radiación , Proteínas Recombinantes/metabolismo , Factores de Empalme Serina-Arginina/fisiología , Especificidad de la Especie
7.
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
8.
Proc Natl Acad Sci U S A ; 113(16): E2306-15, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044077

RESUMEN

Current approaches in tissue engineering are geared toward generating tissue-specific stem cells. Given the complexity and heterogeneity of tissues, this approach has its limitations. An alternate approach is to induce terminally differentiated cells to dedifferentiate into multipotent proliferative cells with the capacity to regenerate all components of a damaged tissue, a phenomenon used by salamanders to regenerate limbs. 5-Azacytidine (AZA) is a nucleoside analog that is used to treat preleukemic and leukemic blood disorders. AZA is also known to induce cell plasticity. We hypothesized that AZA-induced cell plasticity occurs via a transient multipotent cell state and that concomitant exposure to a receptive growth factor might result in the expansion of a plastic and proliferative population of cells. To this end, we treated lineage-committed cells with AZA and screened a number of different growth factors with known activity in mesenchyme-derived tissues. Here, we report that transient treatment with AZA in combination with platelet-derived growth factor-AB converts primary somatic cells into tissue-regenerative multipotent stem (iMS) cells. iMS cells possess a distinct transcriptome, are immunosuppressive, and demonstrate long-term self-renewal, serial clonogenicity, and multigerm layer differentiation potential. Importantly, unlike mesenchymal stem cells, iMS cells contribute directly to in vivo tissue regeneration in a context-dependent manner and, unlike embryonic or pluripotent stem cells, do not form teratomas. Taken together, this vector-free method of generating iMS cells from primary terminally differentiated cells has significant scope for application in tissue regeneration.


Asunto(s)
Azacitidina/farmacología , Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Células Cultivadas , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Especificidad de Órganos/fisiología
9.
J Immunol ; 196(5): 2132-44, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26843326

RESUMEN

Vitamin A has essential but largely unexplained roles in regulating lymphopoiesis. We have previously shown that retinoic acid receptor (RAR) γ-deficient mice have hematopoietic defects, some phenotypes of which were microenvironment induced. Bone marrow (BM) microenvironment cells identified by either their expression of nestin (Nes) or osterix (Osx) have previously been shown to have roles in regulating lymphopoiesis. We therefore conditionally deleted Rarγ in Nes- or Osx-expressing microenvironment cells. Osx cell-specific deletion of Rarγ had no impact on hematopoiesis. In contrast, deletion of Rarγ in Nes-expressing cells resulted in reductions in peripheral blood B cells and CD4(+) T cells, accompanied by reductions of immature PreB cells in BM. The mice lacking Rarγ in Nes-expressing cells also had smaller thymi, with reductions in double-negative 4 T cell precursors, accompanied by reduced numbers of both TCRß(low) immature single-positive CD8(+) cells and double-positive T cells. In the thymus, Nes expression was restricted to thymic stromal cells that expressed cerebellar degeneration-related Ag 1 and lacked expression of epithelial cell adhesion molecule. These cells expressed platelet-derived growth factor α and high transcript levels of Rars, Cxcl12, and stem cell factor (Scf). Short-term treatment of mice with all-trans retinoic acid resulted in increased PreB lymphopoiesis in BM and an increase in thymic double-negative 4 T cells, inverse to that observed upon Nes cell-specific deletion of Rarγ. Collectively, these studies show that RARγ is a regulator of B and T lymphopoiesis via Nes-expressing cells in the BM and thymic microenvironments, respectively.


Asunto(s)
Linfocitos B/citología , Microambiente Celular/inmunología , Linfopoyesis/inmunología , Receptores de Ácido Retinoico/inmunología , Linfocitos T/citología , Animales , Linfocitos B/inmunología , Médula Ósea/inmunología , Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Inmunofenotipificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nestina/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Timo/inmunología , Receptor de Ácido Retinoico gamma
10.
PLoS Genet ; 11(4): e1005160, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25859855

RESUMEN

RECQL4 mutations are associated with Rothmund Thomson Syndrome (RTS), RAPADILINO Syndrome and Baller-Gerold Syndrome. These patients display a range of benign skeletal abnormalities such as low bone mass. In addition, RTS patients have a highly increased incidence of osteosarcoma (OS). The role of RECQL4 in normal adult bone development and homeostasis is largely uncharacterized and how mutation of RECQL4 contributes to OS susceptibility is not known. We hypothesised that Recql4 was required for normal skeletal development and both benign and malignant osteoblast function, which we have tested in the mouse. Recql4 deletion in vivo at the osteoblastic progenitor stage of differentiation resulted in mice with shorter bones and reduced bone volume, assessed at 9 weeks of age. This was associated with an osteoblast intrinsic decrease in mineral apposition rate and bone formation rate in the Recql4-deficient cohorts. Deletion of Recql4 in mature osteoblasts/osteocytes in vivo, however, did not cause a detectable phenotype. Acute deletion of Recql4 in primary osteoblasts or shRNA knockdown in an osteoblastic cell line caused failed proliferation, accompanied by cell cycle arrest, induction of apoptosis and impaired differentiation. When cohorts of animals were aged long term, the loss of Recql4 alone was not sufficient to initiate OS. We then crossed the Recql4fl/fl allele to a fully penetrant OS model (Osx-Cre p53fl/fl). Unexpectedly, the Osx-Cre p53fl/flRecql4fl/fl (dKO) animals had a significantly increased OS-free survival compared to Osx-Cre p53fl/fl or Osx-Cre p53fl/flRecql4fl/+ (het) animals. The extended survival was explained when the Recql4 status in the tumors that arose was assessed, and in no case was there complete deletion of Recql4 in the dKO OS. These data provide a mechanism for the benign skeletal phenotypes of RECQL4 mutation syndromes. We propose that tumor suppression and osteosarcoma susceptibility are most likely a function of mutant, not null, alleles of RECQL4.


Asunto(s)
Neoplasias Óseas/genética , Osteoblastos/metabolismo , Osteogénesis , Osteosarcoma/genética , RecQ Helicasas/metabolismo , Animales , Neoplasias Óseas/metabolismo , Proliferación Celular , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/metabolismo , RecQ Helicasas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Stem Cells ; 33(9): 2838-49, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26033476

RESUMEN

The tyrosine kinase receptor, EphB4, mediates cross-talk between stromal and hematopoietic populations during bone remodeling, fracture repair and arthritis, through its interactions with the ligand, ephrin-B2. This study demonstrated that transgenic EphB4 mice (EphB4 Tg), over-expressing EphB4 under the control of collagen type-1 promoter, exhibited higher frequencies of osteogenic cells and hematopoietic stem/progenitor cells (HSC), correlating with a higher frequency of long-term culture-initiating cells (LTC-IC), compared with wild type (WT) mice. EphB4 Tg stromal feeder layers displayed a greater capacity to support LTC-IC in vitro, where blocking EphB4/ephrin-B2 interactions decreased LTC-IC output. Similarly, short hairpin RNA-mediated EphB4 knockdown in human bone marrow stromal cells reduced their ability to support high ephrin-B2 expressing CD34(+) HSC in LTC-IC cultures. Notably, irradiated EphB4 Tg mouse recipients displayed enhanced bone marrow reconstitution capacity and enhanced homing efficiency of transplanted donor hematopoietic stem/progenitor cells relative to WT controls. Studies examining the expression of hematopoietic supportive factors produced by stromal cells indicated that CXCL12, Angiopoietin-1, IL-6, FLT-3 ligand, and osteopontin expression were more highly expressed in EphB4 Tg stromal cells compared with WT controls. These findings indicate that EphB4 facilitates stromal-mediated support of hematopoiesis, and constitute a novel component of the HSC niche.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Receptor EphB4/biosíntesis , Secuencia de Aminoácidos , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Receptor EphB4/genética , Células del Estroma/metabolismo
12.
Br J Haematol ; 164(2): 280-5, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24383846

RESUMEN

Vitamin A and its derivatives (retinoids) are important regulators of haematopoiesis, acting via retinoic acid receptors (RARs). Epidemiological studies indicated an association of vitamin A deficiency with anaemia in humans. To define the requirements of RARs in erythropoiesis, we evaluated erythroid parameters in RAR germ-line deficient and conditional knock out mice with erythroid specific deletion of RARs. Adult RARγ(-/-) mice were anaemic, however, Epor-Cre Rara(fl/fl) , Epor-Cre Rarg(fl/fl) and Epor-Cre Rara(fl/fl) g(fl/fl) mice were normal, indicating a lack of an erythroid intrinsic RAR function. Therefore, erythroid-specific RAR function is dispensable for erythropoiesis and RARγ plays an erythroid extrinsic role in erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Receptores de Ácido Retinoico/fisiología , Animales , Células de la Médula Ósea/metabolismo , Eritroblastos/metabolismo , Inmunofenotipificación , Ratones , Ratones Noqueados , Fenotipo , Receptores de Eritropoyetina/genética , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Receptor de Ácido Retinoico gamma
13.
Haematologica ; 99(1): 163-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23935020

RESUMEN

Multiple myeloma is an incurable hematologic cancer characterized by the clonal proliferation of malignant plasma cells within the bone marrow. Numerous studies suggest that the myeloma plasma cells occupy and alter the stromal tissue of the bone marrow as a means of enhancing their survival and growth. However, the nature and magnitude of the changes to the stromal cell tissue remain to be determined. In this study, we used mesenchymal stromal cell and osteoblast-related cell surface marker expression (STRO-1 and alkaline phosphatase, respectively) and flow cytometry to enumerate mesenchymal stromal cell and osteoblast numbers in bone marrow recovered from myeloma patients at the time of diagnosis. Using this approach, we identified an increase in the number of STRO-1 positive colony forming mesenchymal stromal cells and a concomitant decrease in alkaline phophatase osteoblasts. Notably, this increase in mesenchymal stromal cell numbers correlated closely with plasma cell burden at the time of diagnosis. In addition, in comparison with the osteoblast population, the STRO-1+ mesenchymal stromal cell population was found to express higher levels of plasma cell- and osteoclast-activating factors, including RANKL and IL-6, providing a mechanism by which an increase in mesenchymal stromal cells may promote and aid the progression of myeloma. Importantly, these findings were faithfully replicated in the C57BL/KaLwRij murine model of myeloma, suggesting that this model may present a unique and clinically relevant system in which to identify and therapeutically modulate the bone microenvironment and, in turn, alter the progression of myeloma disease.


Asunto(s)
Médula Ósea/metabolismo , Médula Ósea/patología , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/metabolismo , Microambiente Tumoral , Animales , Antígenos de Superficie/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Inmunofenotipificación , Linfocinas/metabolismo , Ratones , Gammopatía Monoclonal de Relevancia Indeterminada/metabolismo , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/genética , Células Plasmáticas/patología , Índice de Severidad de la Enfermedad , Ensayo de Tumor de Célula Madre
14.
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.

15.
Am J Pathol ; 180(2): 811-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155108

RESUMEN

Bone marrow (BM) fibrosis is a feature of severe hyperparathyroidism. Consistent with this observation, mice expressing constitutively active parathyroid hormone (PTH)/PTH-related peptide receptors (PPR) in osteoblasts (PPR*Tg) display BM fibrosis. To obtain insight into the nature of BM fibrosis in such a model, a double-mutant mouse expressing constitutively active PPR and green fluorescent protein (GFP) under the control of the type I collagen promoter (PPR*Tg/GFP) was generated. Confocal microscopy and flow cytometry revealed the presence of a cell population expressing GFP (GFP(+)) that was also positive for the hematopoietic marker CD45 in the BM of both PPR*Tg/GFP and control animals. This cell population was expanded in PPR*Tg/GFP. The existence of cells expressing both type I collagen and CD45 in the adult BM was confirmed by IHC and fluorescence-activated cell sorting. An analysis of total RNA extracted from sorted GFP(+)CD45(+) cells showed that these cells produced type I collagen and PTH/PTH-related peptide receptor and receptor activator for NF-κB mRNAs, further supporting their features of being both mesenchymal and hematopoietic lineages. Similar cells, known as fibrocytes, are also present in pathological fibroses. Our findings, thus, indicate that the BM is a permissive microenvironment for the differentiation of fibrocyte-like cells and raise the possibility that these cells could contribute to the pathogenesis of BM fibrosis.


Asunto(s)
Biomarcadores/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mielofibrosis Primaria/patología , Animales , Médula Ósea/metabolismo , Diferenciación Celular , Colágeno Tipo I , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Mielofibrosis Primaria/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
16.
Blood ; 117(21): 5631-42, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21421837

RESUMEN

Erythropoietin (Epo) has been used in the treatment of anemia resulting from numerous etiologies, including renal disease and cancer. However, its effects are controversial and the expression pattern of the Epo receptor (Epo-R) is debated. Using in vivo lineage tracing, we document that within the hematopoietic and mesenchymal lineage, expression of Epo-R is essentially restricted to erythroid lineage cells. As expected, adult mice treated with a clinically relevant dose of Epo had expanded erythropoiesis because of amplification of committed erythroid precursors. Surprisingly, we also found that Epo induced a rapid 26% loss of the trabecular bone volume and impaired B-lymphopoiesis within the bone marrow microenvironment. Despite the loss of trabecular bone, hematopoietic stem cell populations were unaffected. Inhibition of the osteoclast activity with bisphosphonate therapy blocked the Epo-induced bone loss. Intriguingly, bisphosphonate treatment also reduced the magnitude of the erythroid response to Epo. These data demonstrate a previously unrecognized in vivo regulatory network coordinating erythropoiesis, B-lymphopoiesis, and skeletal homeostasis. Importantly, these findings may be relevant to the clinical application of Epo.


Asunto(s)
Linfocitos B/metabolismo , Médula Ósea/efectos de los fármacos , Huesos/metabolismo , Eritropoyesis/fisiología , Eritropoyetina/farmacología , Homeostasis , Linfopoyesis/fisiología , Animales , Médula Ósea/metabolismo , Remodelación Ósea/fisiología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Eritroblastos/metabolismo , Citometría de Flujo , Expresión Génica , Humanos , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Bazo/citología , Bazo/metabolismo
17.
Proc Natl Acad Sci U S A ; 107(32): 14229-34, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20616003

RESUMEN

MicroRNAs influence hematopoietic differentiation, but little is known about their effects on the stem cell state. Here, we report that the microRNA processing enzyme Dicer is essential for stem cell persistence in vivo and a specific microRNA, miR-125a, controls the size of the stem cell population by regulating hematopoietic stem/progenitor cell (HSPC) apoptosis. Conditional deletion of Dicer revealed an absolute dependence for the multipotent HSPC population in a cell-autonomous manner, with increased HSPC apoptosis in mutant animals. An evolutionarily conserved microRNA cluster containing miR-99b, let-7e, and miR-125a was preferentially expressed in long-term hematopoietic stem cells. MicroRNA miR-125a alone was capable of increasing the number of hematopoietic stem cells in vivo by more than 8-fold. This result was accomplished through a differentiation stage-specific reduction of apoptosis in immature hematopoietic progenitors, possibly through targeting multiple proapoptotic genes. Bak1 was directly down-regulated by miR-125a and expression of a 3'UTR-less Bak1 blocked miR-125a-induced hematopoietic expansion in vivo. These data demonstrate cell-state-specific regulation by microRNA and identify a unique microRNA functioning to regulate the stem cell pool size.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Endorribonucleasas/fisiología , Células Madre Hematopoyéticas/citología , MicroARNs/fisiología , Animales , Apoptosis/genética , Recuento de Células , Diferenciación Celular , Regulación hacia Abajo , Homeostasis , Ratones , Datos de Secuencia Molecular , Ribonucleasa III , Proteína Destructora del Antagonista Homólogo bcl-2/genética
18.
J Exp Med ; 203(5): 1283-93, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16682494

RESUMEN

Hematopoietic stem cells (HSCs) sustain lifelong production of all blood cell types through finely balanced divisions leading to self-renewal and differentiation. Although several genes influencing HSC self-renewal have been identified, to date no gene has been described that, when activated, enhances HSC self-renewal and, when inactivated [corrected] promotes HSC differentiation. We observe that the retinoic acid receptor (RAR)gamma is selectively expressed in primitive hematopoietic precursors and that the bone marrow of RARgamma knockout mice exhibit markedly reduced numbers of HSCs associated with increased numbers of more mature progenitor cells compared with wild-type mice. In contrast, RARalpha is widely expressed in hematopoietic cells, but RARalpha knockout mice do not exhibit any HSC or progenitor abnormalities. Primitive hematopoietic precursors overexpressing RARalpha differentiate predominantly to granulocytes in short-term culture, whereas those overexpressing RARgamma exhibit a much more undifferentiated phenotype. Furthermore, loss of RARgamma abrogated the potentiating effects of all-trans retinoic acid on the maintenance of HSCs in ex vivo culture. Finally, pharmacological activation of RARgamma ex vivo promotes HSC self-renewal, as demonstrated by serial transplant studies. We conclude that the RARs have distinct roles in hematopoiesis and that RARgamma is a critical physiological and pharmacological regulator of the balance between HSC self-renewal and differentiation.


Asunto(s)
Médula Ósea/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Receptores de Ácido Retinoico/metabolismo , Animales , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Granulocitos/citología , Granulocitos/fisiología , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Ratones , Receptores de Ácido Retinoico/genética , Tretinoina/farmacología , Receptor de Ácido Retinoico gamma
19.
Nat Cell Biol ; 7(2): 172-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15654333

RESUMEN

Haematopoietic stem cells (HSCs) are capable of shifting from a state of relative quiescence under homeostatic conditions to rapid proliferation under conditions of stress. The mechanisms that regulate the relative quiescence of stem cells and its association with self-renewal are unclear, as is the contribution of molecular regulators of the cell cycle to these decisions. Understanding the mechanisms that govern these transitions will provide important insights into cell-cycle regulation of HSCs and possible therapeutic approaches to expand HSCs. We have investigated the role of two negative regulators of the cell cycle, p27(Kip1) and MAD1, in controlling this transition. Here we show that Mad1(-/-)p27(Kip1-/-) bone marrow has a 5.7-fold increase in the frequency of stem cells, and surprisingly, an expanded pool of quiescent HSCs. However, Mad1(-/-)p27(Kip1-/-) stem cells exhibit an enhanced proliferative response under conditions of stress, such as cytokine stimulation in vitro and regeneration of the haematopoietic system after ablation in vivo. Together these data demonstrate that the MYC-antagonist MAD1 and cyclin-dependent kinase inhibitor p27(Kip1) cooperate to regulate the self-renewal and differentiation of HSCs in a context-dependent manner.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Fosfoproteínas/fisiología , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Trasplante de Médula Ósea , Diferenciación Celular , División Celular , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Citocinas/farmacología , Fluorouracilo/farmacología , Ratones , Ratones Mutantes , Proteínas Nucleares , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA