Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(19): e108536, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-35924455

RESUMEN

During development, hematopoietic stem cells (HSCs) are produced from the hemogenic endothelium and will expand in a transient hematopoietic niche. Prostaglandin E2 (PGE2) is essential during vertebrate development and HSC specification, but its precise source in the embryo remains elusive. Here, we show that in the zebrafish embryo, PGE2 synthesis genes are expressed by distinct stromal cell populations, myeloid (neutrophils, macrophages), and endothelial cells of the caudal hematopoietic tissue. Ablation of myeloid cells, which produce the PGE2 precursor prostaglandin H2 (PGH2), results in loss of HSCs in the caudal hematopoietic tissue, which could be rescued by exogeneous PGE2 or PGH2 supplementation. Endothelial cells contribute by expressing the PGH2 import transporter slco2b1 and ptges3, the enzyme converting PGH2 into PGE2. Of note, differential niche cell expression of PGE2 biosynthesis enzymes is also observed in the mouse fetal liver. Taken altogether, our data suggest that the triad composed of neutrophils, macrophages, and endothelial cells sequentially and synergistically contributes to blood stem cell expansion during vertebrate development.


Asunto(s)
Hemangioblastos , Pez Cebra , Animales , Dinoprostona/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Prostaglandina H2/metabolismo
2.
Development ; 149(19)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36168784

RESUMEN

Hematopoietic stem and progenitor cells emerge from the aorta and migrate to the caudal hematopoietic tissue (CHT) of zebrafish larvae, the hematopoietic equivalent of the mammalian fetal liver, for their proliferation and differentiation. We previously reported that somite-derived stromal cells were a key component of the CHT niche. Here, we found that the cell adhesion protein Protocadherin 18a (Pcdh18a) is expressed in the stromal cell progenitors (SCPs) emigrating from somites toward the future CHT. Deletion of most of the Pcdh18a intracellular domain caused a decrease in the number of SCPs, the directionality of their migration, and the cell-contact mediated repulsion that normally occurs between migrating SCPs. These defects were followed by abnormal morphogenesis of the venous plexus that forms the CHT framework, and the inability of the CHT to function as a niche for hematopoietic stem and progenitor cells. Finally, we found that the extracellular domain of Pcdh18a mediates trans heterophilic adhesion of stromal cells to endothelial cells in vivo and thereby the reticular versus perivascular fate of SCPs. Thus, Pcdh18a expression in SCPs is essential for the proper development of the hematopoietic niche.


Asunto(s)
Células Madre Hematopoyéticas , Pez Cebra , Animales , Células Endoteliales/metabolismo , Mamíferos , Protocadherinas , Nicho de Células Madre , Células del Estroma
3.
Pharm Biol ; 61(1): 768-778, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37148130

RESUMEN

CONTEXT: 5-Fluorouracil (5-FU)-injured stromal cells may cause chronic bone marrow suppression; however, the underlying mechanism remains unclear. Angelica sinensis polysaccharide (ASP), the main biologically active ingredient of the Chinese herb, Angelica sinensis (Oliv.) Diels (Apiaceae), may enrich the blood and promote antioxidation. OBJECTIVE: This study investigated the protective antioxidative effects of ASP on perivascular mesenchymal progenitors (PMPs) and their interactions with hematopoietic cells. MATERIALS AND METHODS: PMPs were dissociated from C57BL/6 mouse femur and tibia and were subsequently divided into the control, ASP (0.1 g/L), 5-FU (0.025 g/L), and 5-FU + ASP (pre-treatment with 0.1 g/L ASP for 6 h, together with 0.025 g/L 5-FU) then cultured for 48 h. Hematopoietic cells were co-cultured on these feeder layers for 24 h. Cell proliferation, senescence, apoptosis, and oxidative indices were detected, along with stromal osteogenic and adipogenic differentiation potentials. Intercellular and intracellular signaling was analyzed by real-time quantitative reverse transcription polymerase chain reaction and Western blotting. RESULTS: ASP ameliorated the reactive oxygen species production/scavenge balance in PMPs; improved osteogenic differentiation; increased SCF, CXCL12, VLA-4/VCAM-1, ICAM-1/LFA1, and TPO/MPL, Ang-1/Tie-2 gene expression. Further, the ASP-treated feeder layer alleviated hematopoietic cells senescence (from 21.9 ± 1.47 to 12.1 ± 1.13); decreased P53, P21, p-GSK-3ß, ß-catenin and cyclin-D1 protein expression, and increased glycogen synthase kinase (GSK)-3ß protein expression in co-cultured hematopoietic cells. DISCUSSION AND CONCLUSIONS: ASP delayed oxidative stress-induced premature senescence of 5-FU-treated feeder co-cultured hematopoietic cells via down-regulation of overactivated Wnt/ß-catenin signaling. These findings provide a new strategy for alleviating myelosuppressive stress.


Asunto(s)
Angelica sinensis , Células Madre Mesenquimatosas , Ratones , Animales , beta Catenina , Glucógeno Sintasa Quinasa 3 beta , Osteogénesis , Ratones Endogámicos C57BL , Estrés Oxidativo , Antioxidantes/farmacología , Vía de Señalización Wnt , Fluorouracilo/toxicidad , Polisacáridos/farmacología
4.
Development ; 146(24)2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31784462

RESUMEN

Stem cell compartments in metazoa get regulated by systemic factors as well as local stem cell niche-derived factors. However, the mechanisms by which systemic signals integrate with local factors in maintaining tissue homeostasis remain unclear. Employing the Drosophila lymph gland, which harbors differentiated blood cells, and stem-like progenitor cells and their niche, we demonstrate how a systemic signal interacts and harmonizes with local factor/s to achieve cell type-specific tissue homeostasis. Our genetic analyses uncovered a novel function of Lar, a receptor protein tyrosine phosphatase. Niche-specific loss of Lar leads to upregulated insulin signaling, causing increased niche cell proliferation and ectopic progenitor differentiation. Insulin signaling assayed by PI3K activation is downregulated after the second instar larval stage, a time point that coincides with the appearance of Lar in the hematopoietic niche. We further demonstrate that Lar physically associates with InR and serves as a negative regulator for insulin signaling in the Drosophila larval hematopoietic niche. Whether Lar serves as a localized invariable negative regulator of systemic signals such as insulin in other stem cell niches remains to be explored.


Asunto(s)
Proteínas de Drosophila/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Homeostasis/genética , Insulina/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/fisiología , Nicho de Células Madre/genética , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Proliferación Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Embrión no Mamífero , Células Madre Hematopoyéticas/fisiología , Unión Proteica , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor de Insulina/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Transducción de Señal/fisiología
5.
Int J Mol Sci ; 23(13)2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35806290

RESUMEN

Hematopoietic stem cells (HSCs) reside in a specialized microenvironment in a peculiar anatomic location which regulates the maintenance of stem cells and controls its functions. Recent scientific progress in experimental technologies have enabled the specific detection of epigenetic factors responsible for the maintenance and quiescence of the hematopoietic niche, which has improved our knowledge of regulatory mechanisms. The aberrant role of RNA-binding proteins and their impact on the disruption of stem cell biology have been reported by a number of recent studies. Despite recent modernization in hematopoietic microenvironment research avenues, our comprehension of the signaling mechanisms and interactive pathways responsible for integration of the hematopoietic niche is still limited. In the past few decades, zebrafish usage with regards to exploratory studies of the hematopoietic niche has expanded our knowledge for deeper understanding of novel cellular interactions. This review provides an update on the functional roles of different genetic and epigenetic factors and molecular signaling events at different sections of the hematopoietic microenvironment. The explorations of different molecular approaches and interventions of latest web-based tools being used are also outlined. This will help us to get more mechanistic insights and develop therapeutic options for the malignancies.


Asunto(s)
Nicho de Células Madre , Pez Cebra , Animales , Comunicación Celular , Biología Computacional , Células Madre Hematopoyéticas/metabolismo
6.
FASEB J ; 34(4): 5435-5452, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086861

RESUMEN

Osteolineage cell-derived extracellular vesicles (EVs) play a regulatory role in hematopoiesis and have been shown to promote the ex vivo expansion of human hematopoietic stem and progenitor cells (HSPCs). Here, we demonstrate that EVs from different human osteolineage sources do not have the same HSPC expansion promoting potential. Comparison of stimulatory and non-stimulatory osteolineage EVs by next-generation sequencing and mass spectrometry analyses revealed distinct microRNA and protein signatures identifying EV-derived candidate regulators of ex vivo HSPC expansion. Accordingly, the treatment of umbilical cord blood-derived CD34+ HSPCs with stimulatory EVs-altered HSPC transcriptome, including genes with known roles in cell proliferation. An integrative bioinformatics approach, which connects the HSPC gene expression data with the candidate cargo in stimulatory EVs, delineated the potentially targeted biological functions and pathways during hematopoietic cell expansion and development. In conclusion, our study gives novel insights into the complex biological role of EVs in osteolineage cell-HSPC crosstalk and promotes the utility of EVs and their cargo as therapeutic agents in regenerative medicine.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Vesículas Extracelulares/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/citología , Osteoblastos/citología , Antígenos CD34/metabolismo , Proliferación Celular , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Humanos , Osteoblastos/metabolismo , Transcriptoma
7.
Proc Natl Acad Sci U S A ; 114(15): 3957-3962, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348230

RESUMEN

RelB is an NF-κB family transcription factor activated in the noncanonical pathway downstream of NF-κB-inducing kinase (NIK) and TNF receptor family members including lymphotoxin-ß receptor (LTßR) and CD40. Early analysis suggested that RelB is required for classical dendritic cell (cDC) development based on a severe reduction of cDCs in Relb-/- mice associated with profound myeloid expansion and perturbations in B and T cells. Subsequent analysis of radiation chimeras generated from wild-type and Relb-/- bone marrow showed that RelB exerts cell-extrinsic actions on some lineages, but it has remained unclear whether the impact of RelB on cDC development is cell-intrinsic or -extrinsic. Here, we reevaluated the role of RelB in cDC and myeloid development using a series of radiation chimeras. We found that there was no cell-intrinsic requirement for RelB for development of most cDC subsets, except for the Notch2- and LTßR-dependent subset of splenic CD4+ cDC2s. These results identify a relatively restricted role of RelB in DC development. Moreover, the myeloid expansion in Relb-/- mice resulted from hematopoietic-extrinsic actions of RelB. This result suggests that there is an unrecognized but critical role for RelB within the nonhematopoietic niche that controls normal myelopoiesis.


Asunto(s)
Células Dendríticas/fisiología , Células Mieloides/fisiología , Factor de Transcripción ReIB/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Sistema Hematopoyético/citología , Sistema Hematopoyético/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Serina-Treonina Quinasas/metabolismo , Bazo/citología , Bazo/metabolismo , Factor de Transcripción ReIB/metabolismo , Quinasa de Factor Nuclear kappa B
8.
Development ; 143(24): 4571-4581, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27965438

RESUMEN

Not all hematopoietic stem cells (HSCs) are alike. They differ in their physical characteristics such as cell cycle status and cell surface marker phenotype, they respond to different extrinsic signals, and they have different lineage outputs following transplantation. The growing body of evidence that supports heterogeneity within HSCs, which constitute the most robust cell fraction at the foundation of the adult hematopoietic system, is currently of great interest and raises questions as to why HSC subtypes exist, how they are generated and whether HSC heterogeneity affects leukemogenesis or treatment options. This Review provides a developmental overview of HSC subtypes during embryonic, fetal and adult stages of hematopoiesis and discusses the possible origins and consequences of HSC heterogeneity.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/clasificación , Células Madre Hematopoyéticas/citología , Animales , Células de la Médula Ósea/citología , Linaje de la Célula/fisiología , Células Endoteliales/citología , Humanos , Nicho de Células Madre
9.
Ann Hematol ; 98(9): 2063-2072, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31312928

RESUMEN

Rigosertib is a novel multi-kinase inhibitor, which has clinical activity towards leukemic progenitor cells of patients with high-risk myelodysplastic syndromes (MDS) after failure or progression on hypomethylating agents. Since the bone marrow microenvironment plays an important role in MDS pathogenesis, we investigated the impact of rigosertib on cellular compartments within the osteo-hematopoietic niche. Healthy C57BL/6J mice treated with rigosertib for 3 weeks showed a mild suppression of hematopoiesis (hemoglobin and red blood cells, both - 16%, p < 0.01; white blood cells, - 34%, p < 0.05; platelets, - 38%, p < 0.05), whereas there was no difference in the number of hematopoietic stem cells in the bone marrow. Trabecular bone mass of the spine was reduced by rigosertib (- 16%, p = 0.05). This was accompanied by a lower trabecular number and thickness (- 6% and - 10%, respectively, p < 0.05), partly explained by the increase in osteoclast number and surface (p < 0.01). Milder effects of rigosertib on bone mass were detected in an MDS mouse model system (NHD13). However, rigosertib did not further aggravate MDS-associated cytopenia in NHD13 mice. Finally, we tested the effects of rigosertib on human mesenchymal stromal cells (MSC) in vitro and demonstrated reduced cell viability at nanomolar concentrations. Deterioration of the hematopoietic supportive capacity of MDS-MSC after rigosertib pretreatment demonstrated by decreased number of colony-forming units, especially in the monocytic lineage, further supports the idea of disturbed crosstalk within the osteo-hematopoietic niche mediated by rigosertib. Thus, rigosertib exerts inhibitory effects on the stromal components of the osteo-hematopoietic niche which may explain the dissociation between anti-leukemic activity and the absence of hematological improvement.


Asunto(s)
Glicina/análogos & derivados , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Síndromes Mielodisplásicos , Nicho de Células Madre/efectos de los fármacos , Sulfonas/farmacología , Animales , Glicina/farmacología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología
10.
Development ; 142(13): 2261-7, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26041767

RESUMEN

Bag of Marbles (Bam) is known to function as a positive regulator of hematopoietic progenitor maintenance in the lymph gland blood cell-forming organ during Drosophila hematopoiesis. Here, we demonstrate a key function for Bam in cells of the lymph gland posterior signaling center (PSC), a cellular domain proven to function as a hematopoietic niche. Bam is expressed in PSC cells, and gene loss-of-function results in PSC overgrowth and disorganization, indicating that Bam plays a crucial role in controlling the proper development of the niche. It was previously shown that Insulin receptor (InR) pathway signaling is essential for proper PSC cell proliferation. We analyzed PSC cell number in lymph glands double-mutant for bam and InR pathway genes, and observed that bam genetically interacts with pathway members in the formation of a normal PSC. The elF4A protein is a translation factor downstream of InR pathway signaling, and functional knockdown of this crucial regulator rescued the bam PSC overgrowth phenotype, further supporting the cooperative function of Bam with InR pathway members. Additionally, we documented that the Retinoblastoma-family protein (Rbf), a proven regulator of cell proliferation, was present in cells of the PSC, with a bam function-dependent expression. By contrast, perturbation of Decapentaplegic or Wingless signaling failed to affect Rbf niche cell expression. Together, these findings indicate that InR pathway-Bam-Rbf functional interactions represent a newly identified means to regulate the correct size and organization of the PSC hematopoietic niche.


Asunto(s)
Tamaño de la Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Madre Hematopoyéticas/citología , Proteína de Retinoblastoma/metabolismo , Somatomedinas/metabolismo , Nicho de Células Madre , Factores de Transcripción/metabolismo , Animales , Recuento de Células , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Epistasis Genética , Factor 4A Eucariótico de Iniciación/genética , Genes de Insecto , Células Madre Hematopoyéticas/metabolismo , Tejido Linfoide/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
11.
Stem Cells ; 35(10): 2150-2159, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28741855

RESUMEN

Bone-forming osteoblasts play critical roles in supporting bone marrow hematopoiesis. Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced PSCs (iPSC), are capable of differentiating into osteoblasts. To determine the capacity of stem cells needed to rescue aberrant skeletal development and bone marrow hematopoiesis in vivo, we used a skeletal complementation model. Mice deficient in Runx2, a master transcription factor for osteoblastogenesis, fail to form a mineralized skeleton and bone marrow. Wild-type (WT) green fluorescent protein (GFP)+ ESCs and yellow fluorescent protein (YFP)+ iPSCs were introduced into Runx2-null blastocyst-stage embryos. We assessed GFP/YFP+ cell contribution by whole-mount fluorescence and histological analysis and found that the proportion of PSCs in the resulting chimeric embryos is directly correlated with the degree of mineralization in the skull. Moreover, PSC contribution to long bones successfully restored bone marrow hematopoiesis. We validated this finding in a separate model with diphtheria toxin A-mediated ablation of hypertrophic chondrocytes and osteoblasts. Remarkably, chimeric embryos harboring as little as 37.5% WT PSCs revealed grossly normal skeletal morphology, suggesting a near-complete rescue of skeletogenesis. In summary, we demonstrate that fractional contribution of PSCs in vivo is sufficient to complement and reconstitute an osteoblast-deficient skeleton and hematopoietic marrow. Further investigation using genetically modified PSCs with conditional loss of gene function in osteoblasts will enable us to address the specific roles of signaling mediators to regulate bone formation and hematopoietic niches in vivo. Stem Cells 2017;35:2150-2159.


Asunto(s)
Osteoblastos/metabolismo , Osteogénesis/fisiología , Células Madre Pluripotentes/metabolismo , Nicho de Células Madre/fisiología , Diferenciación Celular , Humanos
12.
Eur J Haematol ; 100(1): 3-9, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29032607

RESUMEN

This review is a journey of the landmark erythroblast macrophage protein (Emp) discovered in 1994, and it walks chronologically through the progress that has been made in understanding the biological function of this protein. Historically, Emp was the first identified cell attachment molecule and is expressed in both erythroblasts and macrophages and mediates their attachments to form erythroblastic islands. The absence of Emp erythroblasts shows defects in differentiation and enucleation. Emp-deficient macrophages display immature morphology characterized by small sizes, round shapes, and the lack of cytoplasmic projections. Although the primary sequence of Emp has already been determined and its role in both erythroid and macrophage development is well established, there are major gaps in the understanding of its function at the molecular level. Recent studies had implicated its importance in actin cytoskeleton remodeling and cell migration, but the molecular mechanisms are still enigmatic. Previous studies have also demonstrated that downregulation of Emp affects the expression of mitogen-associated protein kinase 1 (MAPK1) and thymoma viral protooncogene (AKT-1) resulting in abnormal cell motility. In this review, we summarize the proposed function of Emp based on previous studies, present scenarios, and its plausible future in translational research.


Asunto(s)
Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Animales , Moléculas de Adhesión Celular/química , Diferenciación Celular , Proteínas del Citoesqueleto/química , Eritroblastos/citología , Eritroblastos/metabolismo , Eritropoyesis/genética , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Transporte de Proteínas
13.
Adv Exp Med Biol ; 1041: 33-53, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29204828

RESUMEN

Hematopoietic stem cells (HSCs) posses the ability to maintain the blood system of an organism from birth to adulthood. The behavior of HSCs is modulated by its microenvironment. During development, HSCs acquire the instructions to self-renew and differentiate into all blood cell fates by passing through several developmental microenvironments. In this chapter, we discuss the signals and cell types that inform HSC decisions throughout ontogeny with a focus on HSC specification, mobilization, migration, and engraftment.


Asunto(s)
Tipificación del Cuerpo/fisiología , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Animales , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Células Madre Hematopoyéticas/citología , Humanos , Factores de Tiempo , Pez Cebra/embriología
14.
Biochem Biophys Res Commun ; 478(4): 1706-12, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27596966

RESUMEN

The expression of extracellular matrix protein periostin (POSTN) was attenuated in Med1(-/-) mouse embryonic fibroblasts (MEFs), which exhibited a decreased capability to support hematopoietic progenitor cells (HPCs) in vitro. When bone marrow (BM) cells were cocultured with mitomycin C-treated Med1(+/+) MEFs, or OP-9 or MS-5 BM stromal cells, in the presence of anti-POSTN antibody, the growth of BM cells and number of long-term culture-initiating cells (LTC-ICs) were attenuated. When BM cells were cocultured with Med1(-/-) MEFs in the presence of recombinant POSTN, the growth of BM cells and the number of LTC-ICs were restored. Moreover, antibody-mediated blockage of stromal cells-derived POSTN markedly reduced the growth and cobblestone formation, a leukemic stem cell feature, of stromal cell-dependent MB-1 myeloblastoma cells. POSTN was expressed both in BM cells and variably in different BM stromal cells. Expression in the latter cells was increased by physical interaction with hematopoietic cells. The receptor for POSTN, integrin αvß3, was expressed abundantly in BM stromal cells. The addition of recombinant POSTN to BM stromal cells induced intracellular signaling downstream of integrin αvß3. These results suggest that stromal cell POSTN supports both normal HPCs and leukemia-initiating cells in vitro, at least in part, indirectly by acting on stromal cells in an autocrine or paracrine manner.


Asunto(s)
Moléculas de Adhesión Celular/genética , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Nicho de Células Madre , Animales , Western Blotting , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma Mieloide/genética , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Int J Mol Sci ; 17(11)2016 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-27827995

RESUMEN

Scid hematopoietic stem cells (HSCs) have an intrinsic defect in their maintenance within the bone marrow (BM) niche which facilitates HSC transplantation without the absolute requirement of prior conditioning. Nevertheless, NOD scid mice have a significantly altered life span due to early development of thymic lymphomas, which compromises the ability to study the long-term fate of exogenous HSCs and their progeny. Here, we present data on the transplantation of HSCs into NOD scid gamma (NSG) mice to achieve long-term engraftment without prior conditioning. We transplanted allogeneic HSCs constitutively expressing the mCherry fluorescent marker into age-matched NSG mice and assessed donor chimerism 6 months post-transplantation. All transplanted NSG mice showed long-term myeloid and lymphoid cell chimerism. Also, in vivo irradiated HSCs showed long-term engraftment, although overall white blood cell (WBC) donor chimerism was lower compared with non-irradiated HSCs. Using this novel NSG transplantation model, we will be able to study the effects of low dose in vivo X-ray exposure on the long-term fate of HSCs, without the requirement of prior radio-ablation of the recipient, and thus leaving the recipient's BM microenvironment uncompromised. In conclusion, we demonstrated for the first time that allogeneic HSCs from a different inbred strain can compete for niches in the BM compartment of NSG mice.


Asunto(s)
Médula Ósea/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/efectos de la radiación , Quimera por Trasplante/inmunología , Animales , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Expresión Génica , Genes Reporteros , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/efectos de la radiación , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Nicho de Células Madre/inmunología , Trasplante Homólogo , Rayos X , Proteína Fluorescente Roja
16.
Int Immunol ; 26(11): 607-18, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24908679

RESUMEN

Bone remodeling and hematopoiesis are interrelated and bone marrow (BM) macrophages are considered to be important for both bone remodeling and maintenance of the hematopoietic niche. We found that NF-κB Rela-deficient chimeric mice, generated by transplanting Rela (-/-) fetal liver cells into lethally irradiated hosts, developed severe osteopenia, reduced lymphopoiesis and enhanced mobilization of hematopoietic stem and progenitor cells when BM cells were completely substituted by Rela-deficient cells. Rela (-/-) hematopoietic stem cells from fetal liver had normal hematopoietic ability, but those harvested from the BM of osteopenic Rela (-/-) chimeric mice had reduced repopulation ability, indicating impairment of the microenvironment for the hematopoietic niche. Osteopenia in Rela (-/-) chimeric mice was due to reduced bone formation, even though osteoblasts differentiated from host cells. This finding indicates impaired functional coupling between osteoblasts and hematopoietic stem cell-derived cells. Rela-deficient BM macrophages exhibited an aberrant inflammatory phenotype, and transplantation with wild-type F4/80(+) BM macrophages recovered bone formation and ameliorated lymphopoiesis in Rela (-/-) chimeric mice. Therefore, RELA in F4/80(+) macrophages is important both for bone homeostasis and for maintaining the hematopoietic niche after lethal irradiation and hematopoietic stem cell transplantation.


Asunto(s)
Hematopoyesis/genética , Macrófagos/metabolismo , Osteogénesis/genética , Nicho de Células Madre/genética , Factor de Transcripción ReIA/deficiencia , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Células Madre Hematopoyéticas , Linfopoyesis/genética , Masculino , Ratones , Ratones Noqueados , Osteoclastos/metabolismo , Factor de Transcripción ReIA/genética , Quimera por Trasplante , Irradiación Corporal Total
17.
Am J Transplant ; 14(4): 820-30, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24592822

RESUMEN

Human cytomegalovirus (HCMV) infection is an important cause of morbidity and mortality among both solid organ and hematopoietic stem cell transplant recipients. Identification of cells throughout the body that can potentially serve as a viral reservoir is essential to dissect mechanisms of cell tropism and latency and to develop novel therapies. Here, we tested and compared the permissivity of liver-, brain-, lung (LNG)- and bone marrow (BM)-derived perivascular mesenchymal stromal cells (MSC) to HCMV infection and their ability to propagate and produce infectious virus. Perivascular MSC isolated from the different organs have in common the expression of CD146 and Stro-1. While all these cells were permissive to HCMV infection, the highest rate of HCMV infection was seen with LNG-MSC, as determined by viral copy number and production of viral particles by these cells. In addition, we showed that, although the supernatants from each of the HCMV-infected cultures contained infectious virus, the viral copy number and the quantity and timing of virus production varied among the various organ-specific MSC. Furthermore, using quantitative polymerase chain reaction, we were able to detect HCMV DNA in BM-MSC isolated from 7 out of 19 healthy, HCMV-seropositive adults, suggesting that BM-derived perivascular stromal cells may constitute an unrecognized natural HCMV reservoir.


Asunto(s)
Infecciones por Citomegalovirus/virología , ADN Viral/genética , Células Madre Mesenquimatosas/virología , Adulto , Médula Ósea/metabolismo , Médula Ósea/virología , Encéfalo/citología , Encéfalo/virología , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Feto/virología , Humanos , Hígado/citología , Hígado/virología , Pulmón/citología , Pulmón/virología , Reacción en Cadena de la Polimerasa , Replicación Viral
18.
J Cutan Pathol ; 41(7): 606-11, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24698478

RESUMEN

Primary osteoma cutis (cutaneous ossification) is an uncommon disease in which there is bone formation within the skin in the absence of a demonstrable pre-existing condition. Osteoma cutis is a chronic and benign condition. We report a case of a 45-year-old man who developed extramedullary acute leukemia with a myeloid immunophenotype (myeloid sarcoma) with its initial presentation within an isolated pre-existing osteoma cutis in the post-auricular scalp without evidence of systemic acute leukemia or chronic myeloid stem cell disorders. The tumor was surgically excised without complications. Four months later, acute leukemia recurred in the contralateral posterior mandible and showed an immunophenotype consistent with acute lymphoblastic leukemia/lymphoma. The patient now has been treated by standard protocols for acute leukemia. The diagnosis of an extramedullary acute leukemia is challenging because of its inconsistent clinical and histopathologic presentations. Extramedullary acute leukemia developing in a pre-existing osteoma cutis is very unusual and has not been previously reported in the literature.


Asunto(s)
Enfermedades Óseas Metabólicas , Neoplasias de Cabeza y Cuello , Osificación Heterotópica , Sarcoma Mieloide , Enfermedades Cutáneas Genéticas , Neoplasias Cutáneas , Enfermedad Aguda , Enfermedades Óseas Metabólicas/patología , Enfermedades Óseas Metabólicas/cirugía , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Masculino , Osificación Heterotópica/patología , Osificación Heterotópica/cirugía , Sarcoma Mieloide/patología , Sarcoma Mieloide/cirugía , Enfermedades Cutáneas Genéticas/patología , Enfermedades Cutáneas Genéticas/cirugía , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/cirugía
19.
Methods Mol Biol ; 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38507212

RESUMEN

Maintenance of hematopoietic stem cell (HSC) function is an orchestrated event between multiple cell types, and crosstalk between these cell types is an essential part of HSC regulation. Among the cell groups of the niche involved in this process are a group of bone-resident macrophages known as osteomacs (OM). Previously, it was demonstrated that OM and osteoblasts contained within neonatal calvarial cells are critical to maintain hematopoietic function. Additionally, interactions between neonatal calvarial cells and megakaryocytes further enhance this hematopoietic activity. In this chapter, we explore one such interaction involving OM and osteoblasts in the hematopoietic niche. We describe a protocol to isolate OM from both neonatal and adult mice, and subsequently use colony-forming assays to demonstrate their interaction with osteoblasts in maintaining HSC function.

20.
Acta Naturae ; 16(2): 4-21, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39188265

RESUMEN

As a model organism, the fruit fly (Drosophila melanogaster) has assumed a leading position in modern biological research. The Drosophila genetic system has a number of advantages making it a key model in investigating the molecular mechanisms of metazoan developmental processes. Over the past two decades, significant progress has been made in understanding the molecular mechanisms regulating Drosophila hematopoiesis. This review discusses the major advances in investigating the molecular mechanisms involved in maintaining the population of multipotent progenitor cells and their differentiation into mature hemocytes in the hematopoietic organ of the Drosophila larva. The use of the Drosophila hematopoietic organ as a model system for hematopoiesis has allowed to characterize the complex interactions between signaling pathways and transcription factors in regulating the maintenance and differentiation of progenitor cells through the signals from the hematopoietic niche, autocrine and paracrine signals, and the signals emanated by differentiated cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA