Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Immunol ; 206(7): 1549-1560, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33637617

RESUMEN

Outside-in integrin signaling regulates cell fate decisions in a variety of cell types, including hematopoietic stem cells (HSCs). Our earlier published studies showed that interruption of periostin (POSTN) and integrin-αv (ITGAV) interaction induces faster proliferation in HSCs with developmental stage-dependent functional effects. In this study, we examined the role of POSTN-ITGAV axis in lymphohematopoietic activity in spleen that hosts a rare population of HSCs, the functional regulation of which is not clearly known. Vav-iCre-mediated deletion of Itgav in the hematopoietic system led to higher proliferation rates, resulting in increased frequency of primitive HSCs in the adult spleen. However, in vitro CFU-C assays demonstrated a poorer differentiation potential following Itgav deletion. This also led to a decrease in the white pulp area with a significant decline in the B cell numbers. Systemic deletion of its ligand, POSTN, phenocopied the effects noted in Vav-Itgav-/- mice. Histological examination of Postn-deficient spleen also showed an increase in the spleen trabecular areas. Importantly, these are the myofibroblasts of the trabecular and capsular areas that expressed high levels of POSTN within the spleen tissue. In addition, vascular smooth muscle cells also expressed POSTN. Through CFU-S12 assays, we showed that hematopoietic support potential of stroma in Postn-deficient splenic hematopoietic niche was defective. Overall, we demonstrate that POSTN-ITGAV interaction plays an important role in spleen lymphohematopoiesis.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Madre Hematopoyéticas/fisiología , Integrina alfa5/metabolismo , Linfocitos/fisiología , Miocitos del Músculo Liso/fisiología , Miofibroblastos/fisiología , Bazo/inmunología , Animales , Moléculas de Adhesión Celular/genética , Proliferación Celular , Técnicas de Silenciamiento del Gen , Hematopoyesis , Integrina alfa5/genética , Ratones , Ratones Noqueados , Transducción de Señal , Nicho de Células Madre
2.
Adv Exp Med Biol ; 1132: 163-176, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31037634

RESUMEN

Stem cell function is regulated by a huge repertoire of external cues along with stem cell intrinsic genetic and epigenetic factors. These interactions come through a variety of cell adhesion receptors, of which integrins are one of the most important classes. They interact with extracellular matrix (ECM) components and various bound proteins. Apart from inside-out signaling through which integrins ensure that the cells are stably bound to the ECM, outside-in integrin signaling, through binding to a variety of ligands, play important roles in cell fate decisions. Periostin is one such ligand whose role in functional regulation of stem cells is emerging due to its wide expression profile. In this review, we discuss the recent advancements made in the field.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Integrinas/fisiología , Transducción de Señal , Células Madre/citología , Adhesión Celular , Matriz Extracelular , Humanos
3.
IUBMB Life ; 70(7): 612-624, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29999238

RESUMEN

The hematopoietic system has a very well-studied hierarchy with the long-term (LT) hematopoietic stem cells (HSCs) taking the top position. The pool of quiescent adult LT-HSCs generated during the fetal and early postnatal life acts as a reservoir to supply all the blood cells. Therefore, the maintenance of this stem cell pool is pivotal to maintaining homeostasis in hematopoietic system. It has long been known that external cues, along with the internal genetic factors influence the status of HSCs in the bone marrow (BM). Hypoxia is one such factor that regulates the vascular as well as hematopoietic ontogeny from a very early time point in development. The metabolic outcomes of a hypoxic microenvironment play important roles in functional regulation of HSCs, especially in case of adult BM HSCs. Anaerobic metabolic pathways therefore perform prominent role in meeting energy demands. Increased oxidative pathways on the other hand result in loss of stemness. Recent studies have attributed the functional differences in HSCs across different life stages to their metabolic phenotypes regulated by respective niches. Indicating thus, that various energy production pathways could play distinct role in regulating HSC function at different developmental/physiological states. Here, we review the current status of our understanding over the role that energy production pathways play in regulating HSC stemness. © 2018 IUBMB Life, 70(7):612-624, 2018.


Asunto(s)
Envejecimiento/fisiología , Metabolismo Energético , Células Madre Hematopoyéticas/fisiología , Animales , Células Madre Hematopoyéticas/metabolismo , Humanos , Redes y Vías Metabólicas , Mitocondrias/metabolismo , Oxidación-Reducción
4.
Cytotherapy ; 19(6): 744-755, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28499585

RESUMEN

BACKGROUND AIMS: Myelodysplastic syndromes (MDS) are a group of clonal stem cell disorders affecting the normal hematopoietic differentiation process and leading to abnormal maturation and differentiation of all blood cell lineages. Treatment options are limited, and there is an unmet medical need for effective therapies for patients with severe cytopenias. METHODS: We demonstrate that multipotent adult progenitor cells (MAPC) improve the function of hematopoietic progenitors derived from human MDS bone marrow (BM) by significantly increasing the frequency of primitive progenitors as well as the number of myeloid colonies. RESULTS: This effect was more pronounced in a non-contact culture, indicating the importance of soluble factors produced by the MAPC cells. Moreover, the cells did not stimulate the growth of the abnormal MDS clone, as shown by fluorescent in situ hybridization analysis on BM cells from patients with a known genetic abnormality. We also demonstrate that MAPC cells can provide stromal support for patient-derived hematopoietic cells. When MAPC cells were intravenously injected into a mouse model of MDS, they migrated to the site of injury and increased the hematopoietic function in diseased mice. DISCUSSION: The preclinical studies undertaken here indicate an initial proof of concept for the use of MAPC cell therapy in patients with MDS-related severe and symptomatic cytopenias and should pave the way for further investigation in clinical trials.


Asunto(s)
Células Madre Multipotentes/trasplante , Síndromes Mielodisplásicos/terapia , Adulto , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Femenino , Hematopoyesis , Humanos , Hibridación Fluorescente in Situ , Ratones Endogámicos C57BL
5.
Dev Dyn ; 245(7): 739-50, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26813236

RESUMEN

In most of the mammalian tissues, homeostasis as well as injury repair depend upon a small number of resident adult stem cells. The decline in tissue/organ function in aged organisms has been directly linked with poorly functioning stem cells. Altered function of hematopoietic stem cells (HSCs) is at the center of an aging hematopoietic system, a tissue with high cellular turnover. Poorly engrafting, myeloid-biased HSCs with higher levels of DNA damage accumulation are the hallmark features of an aged hematopoietic system. These cells show a higher proliferation rate than their younger counterparts. It was proposed that quiescence of these cells over long period of time leads to accumulation of DNA damage, eventually resulting in poor function/pathological conditions in hematopoietic system. However, various mouse models with premature aging phenotype also show highly proliferative HSCs. This review examines the evidence that links proliferation of HSCs with aging, which leads to functional changes in the hematopoietic system. Developmental Dynamics 245:739-750, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proliferación Celular/fisiología , Daño del ADN/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular/genética , Daño del ADN/genética , Células Madre Hematopoyéticas/fisiología , Humanos
6.
Blood ; 121(14): 2587-95, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23327927

RESUMEN

Directional migration determines hematopoietic stem/progenitor cell (HSPC) homing, which depends upon the interaction between the chemokine CXCL12 and its receptor CXCR4. CD26 is a widely expressed membrane-bound ectopeptidase that cleaves CXCL12 thereby depleting its chemokine activity. We identified tissue-factor pathway inhibitor (TFPI) as a biological inhibitor of CD26 in murine and human HSPCs. We observed low-level TFPI expression in endothelial cells in the bone marrow (BM), which did not increase following radiation injury. Treatment of HSPCs with TFPI in vitro led to enhanced HSPC migration toward CXCL12, as well as homing and engraftment in the BM upon transplantation. We found that Glypican-3 (GPC3), a heparan sulfate proteoglycan expressed on murine as well as human HSPCs, mediated this effect. TFPI did not affect CD26 activity, migration, or homing of GPC3(-/-) HSPCs, while it affected GPC1(-/-) HSPCs similar to wild-type HSPCs. Moreover, proliferation of GPC3(-/-) but not GPC1(-/-) BM HSPCs was significantly increased, which was associated with a decrease in the primitive HSC pool in BM and an increase in proportion of the circulating HSPCs in the peripheral blood. Hence, we present a novel role for TFPI and GPC3 in regulating HSC homing as well as retention in the BM.


Asunto(s)
Movimiento Celular/fisiología , Dipeptidil Peptidasa 4/metabolismo , Glipicanos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Lipoproteínas/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Células Cultivadas , Quimiotaxis/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Femenino , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
7.
Blood ; 121(5): 781-90, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23243277

RESUMEN

UNLABELLED: Although it is well established that BMP4 plays an important role in the development of hematopoietic system, it is less well understood whether BMP4 affects adult hematopoiesis and how. Here, we describe a novel mechanism by which BMP4 regulates homing of murine as well as human hematopoietic stem/progenitor cells (HSPCs). BMP4 treatment of murine BM derived c-kitLin-Sca-1 (KLS) and CD150CD48-KLS cells for up to 5 days in vitro prevented the culture-induced loss of Integrin-α4 (ITGA4) expression as well as homing. The effect on ITGA4 expression in response to BMP4 is mediated via SMAD-independent phosphorylation of p38 MAPK, which activates microphthalmia-associated transcription factor (MITF), known to induce ITGA4 expression. Elevated ITGA4 expression significantly enhanced HSPC attachment to bone marrow stromal cells, homing and long-term engraftment of the BMP4 treated cells compared with the cells cultured without BMP4. BMP4 also induced expression of ITGA4 on human BM derived Lin-CD34 cells in culture, which was associated with improved homing potential. Thus, BMP4 prevents culture-induced loss of ITGA4 expression on HSPCs in a SMAD-independent manner, resulting in improved homing of cultured HSPCs and subsequent hematopoietic reconstitution. KEY POINTS: Cytokine-induced loss of murine as well as human HSPC homing during ex vivo culture can be prevented by addition of BMP4. In HSPCs, BMP4 directly regulates Integrin-α4 expression through SMAD-independent p38 MAPK-mediated signaling.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/metabolismo , Integrina alfa4/biosíntesis , Proteínas Smad/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/farmacología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Integrina alfa4/genética , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/biosíntesis , Factor de Transcripción Asociado a Microftalmía/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Smad/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Stem Cells ; 32(11): 3012-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25069965

RESUMEN

We recently demonstrated that ex vivo activation of SMAD-independent bone morphogenetic protein 4 (BMP4) signaling in hematopoietic stem/progenitor cells (HSPCs) influences their homing into the bone marrow (BM). Here, we assessed whether alterations in BMP signaling in vivo affects adult hematopoiesis by affecting the BM niche. We demonstrate that systemic inhibition of SMAD-dependent BMP signaling by infusion of the BMP antagonist noggin (NGN) significantly increased CXCL12 levels in BM plasma leading to enhanced homing and engraftment of transplanted HSPCs. Conversely, the infusion of BMP7 but not BMP4, resulted in decreased HSPC homing. Using ST2 cells as an in vitro model of BM niche, we found that incubation with neutralizing anti-BMP4 antibodies, NGN, or dorsomorphin (DM) as well as knockdown of Smad1/5 and Bmp4, all enhanced CXCL12 production. Chromatin immunoprecipitation identified the SMAD-binding element in the CXCL12 promoter to which SMAD4 binds. When deleted, increased CXCL12 promoter activity was observed, and NGN or DM no longer affected Cxcl12 expression. Interestingly, BMP7 infusion resulted in mobilization of only short-term HSCs, likely because BMP7 affected CXCL12 expression only in osteoblasts but not in other niche components. Hence, we describe SMAD-dependent BMP signaling as a novel regulator of CXCL12 production in the BM niche, influencing HSPC homing, engraftment, and mobilization.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/metabolismo , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Nicho de Células Madre , Animales , Proteína Morfogenética Ósea 4/metabolismo , Linaje de la Célula , Movimiento Celular/fisiología , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Receptores CXCR4/metabolismo
9.
Sci Adv ; 10(7): eadh8478, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38363844

RESUMEN

The first definitive hematopoietic progenitors emerge through the process of endothelial-to-hematopoietic transition in vertebrate embryos. With molecular regulators for this process worked out, the role of metabolic pathways used remains unclear. Here, we performed nano-LC-MS/MS-based proteomic analysis and predicted a metabolic switch from a glycolytic to oxidative state upon hematopoietic transition. Mitochondrial activity, glucose uptake, and glycolytic flux analysis supported this hypothesis. Systemic inhibition of lactate dehydrogenase A (LDHA) increased oxygen consumption rate in the hemato-endothelial system and inhibited the emergence of intra-aortic hematopoietic clusters. These findings were corroborated using Tie2-Cre-mediated deletion of Ldha that showed similar effects on hematopoietic emergence. Conversely, stabilization of HIF-1α via inhibition of oxygen-sensing pathway led to decreased oxidative flux and promoted hematopoietic emergence in mid-gestation embryos. Thus, cell-intrinsic regulation of metabolic state overrides oxygenated microenvironment in the aorta to promote a glycolytic metabolic state that is crucial for hematopoietic emergence in mammalian embryos.


Asunto(s)
Células Madre Hematopoyéticas , Proteómica , Animales , Células Madre Hematopoyéticas/metabolismo , Espectrometría de Masas en Tándem , Endotelio Vascular/metabolismo , Hematopoyesis/fisiología , Mamíferos
10.
Adv Healthc Mater ; : e2400256, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669674

RESUMEN

Cancer is indisputably one of the major threats to mankind, and hence the design of new approaches for the improvement of existing therapeutic strategies is always wanted. Herein, the design of a tumor microenvironment-responsive, DNA-based chemodynamic therapy (CDT) nanoagent with dual Fenton reaction centers for targeted cancer therapy is reported. Self-assembly of DNA amphiphile containing copper complex as the hydrophobic Fenton reaction center results in the formation of CDT-active DNAsome with Cu2+-based Fenton catalytic site as the hydrophobic core and hydrophilic ssDNA protrude on the surface. DNA-based surface addressability of the DNAsome is then used for the integration of second Fenton reaction center, which is a peroxidase-mimicking DNAzyme noncovalently loaded with Hemin and Doxorubicin, via DNA hybridization to give a CDT agent having dual Fenton reaction centers. Targeted internalization of the CDT nanoagent and selective generation of •OH inside HeLa cell are also shown. Excellent therapeutic efficiency is observed for the CDT nanoagent both in vitro and in vivo, and the enhanced efficacy is attributed to the combined and synergetic action of CDT and chemotherapy.

11.
Sci Rep ; 14(1): 3542, 2024 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-38346989

RESUMEN

This article reports the development of a microscopy imaging system that gives feasibility for studying spatio-temporal dynamics of physiological activities of alive biological specimens (over entire volume not only for a particular section, i.e., in 4D). The imaging technology facilitates to obtain two image frames of a section of the larger specimen ([Formula: see text]) with different FOVs at different resolutions or magnifications simultaneously in real-time (in addition to recovery of 3D (volume) information). Again, this imaging system addresses the longstanding challenges of housing multiple light sources (6 at the maximum till date) in microscopy (in general) and light sheet fluorescence microscopy (LSFM) (in particular), by using a tuneable pulsed laser source (with an operating wavelength in the range [Formula: see text]-670 nm) in contrast to the conventional CW laser source being adopted for inducing photo-excitation of tagged fluorophores. In the present study, we employ four wavelengths ([Formula: see text] 488 nm, 585 nm, 590 nm, and 594 nm). Our study also demonstrates quantitative characterization of spatio-temporal dynamics (velocity-both amplitude and direction) of organelles (mitochondria) and their mutual correlationships. Mitochondria close to the nucleus (or in clustered cells) are observed to possess a lower degree of freedom in comparison to that at the cellular periphery (or isolated cells). In addition, the study demonstrates real-time observation and recording of the development and growth of all tracheal branches during the entire period ([Formula: see text] min) of embryonic development (Drosophila). The experimental results-with experiments being conducted in various and diversified biological specimens (Drosophila melanogaster, mouse embryo, and HeLa cells)-demonstrate that the study is of great scientific impact both from the aspects of technology and biological sciences.


Asunto(s)
Drosophila melanogaster , Drosophila , Humanos , Animales , Ratones , Células HeLa , Estudios de Tiempo y Movimiento , Microscopía Fluorescente/métodos
12.
STAR Protoc ; 3(4): 101580, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36223268

RESUMEN

Understanding the murine fetal liver (FL) hematopoietic microenvironment, which promotes HSC proliferation, warrants identifying innate relationships between stem cells and the niche. An inclusive study of these cell associations remains elusive. Here, we optimized a protocol to immunolabel HSCs alongside the FL vasculature, a promising niche component. We provide a comprehensive plan from tissue processing, immunohistochemistry, and confocal microscopy, to three-dimensional distance analyses between HSCs and vasculature. This technique can be adapted for achieving congruous outcomes for other cell types. For complete details on the use and execution of this protocol, please refer to Biswas et al. (2020).


Asunto(s)
Células Madre Hematopoyéticas , Hígado , Animales , Ratones , Hígado/metabolismo , Células Madre Hematopoyéticas/metabolismo
13.
iScience ; 25(10): 105171, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36204266

RESUMEN

Interaction with microenvironmental factors is crucial for the regulation of hematopoietic stem cell (HSC) function. Stroma derived factor (SDF)-1α supports HSCs in the quiescent state and is central to the homing of transplanted HSCs. Here, we show that integrin signaling regulates Sdf-1α expression transcriptionally. Systemic deletion of Periostin, an Integrin-αv ligand, showed increased expression of Sdf-1α in bone marrow (BM) niche. Pharmacological inhibition or CRISPR-Cas9-mediated deletion of SRC, resulted in a similar increase in the chemokine expression in vitro. Importantly, systemic SRC-inhibition led to increase in SDF-1α levels in BM plasma. This resulted in a robust increase (14.05 ± 1.22% to 29.11 ± 0.69%) in the homing efficiency of transplanted HSCs. In addition, we observed enhancement in the recovery of blood cell counts following radiation injury, indicating an enhanced hematopoietic function. These results establish a role of SRC-mediated integrin signaling in the transcriptional regulation of Sdf-1α. This mechanism could be harnessed further to improve the hematopoietic function.

14.
J Biol Chem ; 285(7): 4725-31, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20018873

RESUMEN

Hematopoietic stem cells can directly transdifferentiate into hepatocytes because of cellular plasticity, but the molecular basis of transdifferentiation is not known. Here, we show the molecular basis using lineage-depleted oncostatin M receptor beta-expressing (Lin(-)OSMRbeta(+)) mouse bone marrow cells in a hepatic differentiation culture system. Differentiation of the cells was marked by the expression of albumin. Hepatocyte nuclear factor (HNF)-4alpha was expressed and translocated into the nuclei of the differentiating cells. Suppression of its activation in OSM-neutralized culture medium inhibited cellular differentiation. Ectopic expression of full-length HNF4alpha in 32D myeloid cells resulted in decreased myeloid colony-forming potential and increased expression of hepatocyte-specific genes and proteins. Nevertheless, the neohepatocytes produced in culture expressed active P450 enzyme. The obligatory role of HNF4alpha in hepatic differentiation was confirmed by transfecting Lin(-)OSMRbeta(+) cells with dominant negative HNF4alpha in the differentiation culture because its expression inhibited the transcription of the albumin and tyrosine aminotransferase genes. The loss and gain of functional activities strongly suggested that HNF4alpha plays a central role in the transdifferentiation process. For the first time, this report demonstrates the mechanism of transdifferentiation of hematopoietic cells into hepatocytes, in which HNF4alpha serves as a molecular switch.


Asunto(s)
Hematopoyesis/fisiología , Factor Nuclear 4 del Hepatocito/fisiología , Hepatocitos/citología , Hepatocitos/metabolismo , Animales , Western Blotting , Transdiferenciación Celular , Células Cultivadas , Citometría de Flujo , Hematopoyesis/genética , Factor Nuclear 4 del Hepatocito/genética , Inmunohistoquímica , Cinética , Ratones , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Bio Protoc ; 11(16): e4130, 2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34541048

RESUMEN

In vertebrates, hematopoietic stem cells (HSCs) regulate the supply of blood cells throughout the lifetime and help to maintain homeostasis. Due to their long lifespan, genetic integrity is paramount for these cells, and accordingly, a number of stem cell-specific mechanisms are employed. However, HSCs tend to show more DNA damage with increasing age due to an imbalance between proliferation rates and DNA damage responses. The comet assay is the most common and reliable method to study DNA strand breaks at the single-cell level. This procedure is based on the electrophoresis of agarose-embedded lysed cells. Following the electrophoretic mobilization of DNA, it is stained with fluorescent DNA-binding dye. Broken DNA strands migrate based on fragment size and form a tail-like structure called "the comet," whereas intact nuclear DNA remains a part of the head of the comet. Since the alkaline comet assay fails to differentiate between single and double-strand breaks (DSBs), we used a neutral comet assay to quantitate the DSBs in HSCs upon aging and other physiological stresses. The protocol presented here provides procedural details on this highly sensitive, rapid, and cost-effective assay, which can be used for rare populations of cells such as HSCs. Graphical abstract: The neutral comet assay is an extremely useful tool that allows the detection and quantitation of double-strand DNA breaks at the single-cell level. The graphical abstract represents a flowchart for the neutral comet assay procedure.

16.
Cell Rep ; 36(8): 109618, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433017

RESUMEN

Hematopoietic stem and progenitor cell (HSPC) engraftment after transplantation during anticancer treatment depends on support from the recipient bone marrow (BM) microenvironment. Here, by studying physiological homing of fetal HSPCs, we show the critical requirement of balanced local crosstalk within the skeletal niche for successful HSPC settlement in BM. Transgene-induced overproduction of vascular endothelial growth factor (VEGF) by osteoprogenitor cells elicits stromal and endothelial hyperactivation, profoundly impacting the stromal-vessel interface and vascular architecture. Concomitantly, HSPC homing and survival are drastically impaired. Transcriptome profiling, flow cytometry, and high-resolution imaging indicate alterations in perivascular and endothelial cell characteristics, vascular function and cellular metabolism, associated with increased oxidative stress within the VEGF-enriched BM environment. Thus, developmental HSPC homing to bone is controlled by local stromal-vascular integrity and the oxidative-metabolic status of the recipient milieu. Interestingly, irradiation of adult mice also induces stromal VEGF expression and similar osteo-angiogenic niche changes, underscoring that our findings may contribute targets for improving stem cell therapies.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Estrés Oxidativo/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Movimiento Celular/fisiología , Células Cultivadas , Ratones , Nicho de Células Madre/fisiología , Trasplante de Células Madre/métodos
17.
Stem Cell Reports ; 15(2): 340-357, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32735820

RESUMEN

We earlier showed that outside-in integrin signaling through POSTN-ITGAV interaction plays an important role in regulating adult hematopoietic stem cell (HSC) quiescence. Here, we show that Itgav deletion results in increased frequency of phenotypic HSCs in fetal liver (FL) due to faster proliferation. Systemic deletion of Postn led to increased proliferation of FL HSCs, albeit without any loss of stemness, unlike Vav-Itgav-/- HSCs. Based on RNA sequencing analysis of FL and bone marrow HSCs, we predicted the involvement of DNA damage response pathways in this dichotomy. Indeed, proliferative HSCs from Postn-deficient FL tissues showed increased levels of DNA repair, resulting in lesser double-strand breaks. Thus POSTN, with its expression majorly localized in the vascular endothelium of FL tissue, acts as a regulator of stem cell pool size during development. Overall, we demonstrate that the duality of response to proliferation in HSCs is developmental stage dependent and can be correlated with DNA damage responses.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Feto/citología , Células Madre Hematopoyéticas/metabolismo , Integrina alfaV/metabolismo , Hígado/embriología , Transducción de Señal , Animales , Daño del ADN , Reparación del ADN , Endotelio Vascular/metabolismo , Eliminación de Gen , Integrina beta3/metabolismo , Ratones , Ratones Noqueados , Fenotipo
18.
Am J Pathol ; 173(6): 1818-27, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18988804

RESUMEN

Bipotential hepatoblasts differentiate into hepatocytes and cholangiocytes during liver development. It is believed that hepatoblasts originate from endodermal tissue. Here, we provide evidence for the presence of hepatic progenitor cells in the hematopoietic compartment at an early stage of liver development. Flow cytometric analysis showed that at early stages of liver development, approximately 13% of CD45(+) cells express Delta-like protein-1, a marker of hepatoblasts. Furthermore, reverse transcriptase-PCR data suggest that many hepatic genes are expressed in these cells. Cell culture experiments confirmed the hepatic differentiation potential of these cells with the loss of the CD45 marker. We observed that both hematopoietic activity in Delta-like protein-1(+) cells and hepatic activity in CD45(+) cells were high at embryonic day 10.5 and declined thereafter. Clonal analysis revealed that the hematopoietic fraction of fetal liver cells at embryonic day 10.5 gave rise to both hepatic and hematopoietic colonies. The above results suggest a common source of these two functionally distinct cell lineages. In utero transplantation experiments confirmed these results, as green fluorescent protein-expressing CD45(+) cells at the same stage of development yielded functional hepatocytes and hematopoietic reconstitution. Since these cells were unable to differentiate into cytokeratin-19-expressing cholangiocytes, we distinguished them from hepatoblasts. This preliminary study provides hope to correct many liver diseases during prenatal development via transplantation of fetal liver hematopoietic cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Hígado/citología , Hígado/embriología , Animales , Proteínas de Unión al Calcio , Linaje de la Célula , Trasplante de Células , Células Cultivadas , Técnicas de Cocultivo , Femenino , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Hígado/fisiología , Masculino , Ratones , Ratones Transgénicos , Embarazo
19.
PLoS One ; 13(5): e0197046, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29750821

RESUMEN

Multipotent Adult Progenitor Cells (MAPCs) are one potential stem cell source to generate functional hepatocytes or ß-cells. However, human MAPCs have less plasticity than pluripotent stem cells (PSCs), as their ability to generate endodermal cells is not robust. Here we studied the role of 14 transcription factors (TFs) in reprogramming MAPCs to induced endodermal progenitor cells (iENDO cells), defined as cells that can be long-term expanded and differentiated to both hepatocyte- and endocrine pancreatic-like cells. We demonstrated that 14 TF-iENDO cells can be expanded for at least 20 passages, differentiate spontaneously to hepatocyte-, endocrine pancreatic-, gut tube-like cells as well as endodermal tumor formation when grafted in immunodeficient mice. Furthermore, iENDO cells can be differentiated in vitro into hepatocyte- and endocrine pancreatic-like cells. However, the pluripotency TF OCT4, which is not silenced in iENDO cells, may contribute to the incomplete differentiation to mature cells in vitro and to endodermal tumor formation in vivo. Nevertheless, the studies presented here provide evidence that reprogramming of adult stem cells to an endodermal intermediate progenitor, which can be expanded and differentiate to multiple endodermal cell types, might be a valid alternative for the use of PSCs for creation of endodermal cell types.


Asunto(s)
Diferenciación Celular , Endodermo/metabolismo , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/metabolismo , Técnicas de Reprogramación Celular , Endodermo/citología , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología
20.
Stem Cells Dev ; 26(8): 573-584, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27958775

RESUMEN

During ontogeny, fetal liver (FL) acts as a major site for hematopoietic stem cell (HSC) maturation and expansion, whereas HSCs in the adult bone marrow (ABM) are largely quiescent. HSCs in the FL possess faster repopulation capacity as compared with ABM HSCs. However, the molecular mechanism regulating the greater self-renewal potential of FL HSCs has not yet extensively been assessed. Recently, we published RNA sequencing-based gene expression analysis on FL HSCs from 14.5-day mouse embryo (E14.5) in comparison to the ABM HSCs. We reanalyzed these data to identify key transcriptional regulators that play important roles in the expansion of HSCs during development. The comparison of FL E14.5 with ABM HSCs identified more than 1,400 differentially expressed genes. More than 200 genes were shortlisted based on the gene ontology (GO) annotation term "transcription." By morpholino-based knockdown studies in zebrafish, we assessed the function of 18 of these regulators, previously not associated with HSC proliferation. Our studies identified a previously unknown role for tdg, uhrf1, uchl5, and ncoa1 in the emergence of definitive hematopoiesis in zebrafish. In conclusion, we demonstrate that identification of genes involved in transcriptional regulation differentially expressed between expanding FL HSCs and quiescent ABM HSCs, uncovers novel regulators of HSC function.


Asunto(s)
Células Madre Adultas/metabolismo , Células Madre Embrionarias/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hígado/citología , Transcriptoma , Células Madre Adultas/citología , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA