Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO Rep ; 24(5): e55373, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36943011

RESUMEN

Upon ex vivo culture, hematopoietic stem cells (HSCs) quickly lose potential and differentiate into progenitors. The identification of culture conditions that maintain the potential of HSCs ex vivo is therefore of high clinical interest. Here, we demonstrate that the potential of murine and human HSCs is maintained when cultivated for 2 days ex vivo at a pH of 6.9, in contrast to cultivation at the commonly used pH of 7.4. When cultivated at a pH of 6.9, HSCs remain smaller, less metabolically active, less proliferative and show enhanced reconstitution ability upon transplantation compared to HSC cultivated at pH 7.4. HSCs kept at pH 6.9 show an attenuated polyamine pathway. Pharmacological inhibition of the polyamine pathway in HSCs cultivated at pH 7.4 with DFMO mimics phenotypes and potential of HSCs cultivated at pH 6.9. Ex vivo exposure to a pH of 6.9 is therefore a positive regulator of HSC function by reducing polyamines. These findings might improve HSC short-term cultivation protocols for transplantation and gene therapy interventions.


Asunto(s)
Células Madre Hematopoyéticas , Humanos , Ratones , Animales , Células Madre Hematopoyéticas/metabolismo , Concentración de Iones de Hidrógeno
2.
PLoS Biol ; 16(9): e2003389, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30235201

RESUMEN

Hematopoietic stem cells (HSCs) balance self-renewal and differentiation to maintain homeostasis. With aging, the frequency of polar HSCs decreases. Cell polarity in HSCs is controlled by the activity of the small RhoGTPase cell division control protein 42 (Cdc42). Here we demonstrate-using a comprehensive set of paired daughter cell analyses that include single-cell 3D confocal imaging, single-cell transplants, single-cell RNA-seq, and single-cell transposase-accessible chromatin sequencing (ATAC-seq)-that the outcome of HSC divisions is strongly linked to the polarity status before mitosis, which is in turn determined by the level of the activity Cdc42 in stem cells. Aged apolar HSCs undergo preferentially self-renewing symmetric divisions, resulting in daughter stem cells with reduced regenerative capacity and lymphoid potential, while young polar HSCs undergo preferentially asymmetric divisions. Mathematical modeling in combination with experimental data implies a mechanistic role of the asymmetric sorting of Cdc42 in determining the potential of daughter cells via epigenetic mechanisms. Therefore, molecules that control HSC polarity might serve as modulators of the mode of stem cell division regulating the potential of daughter cells.


Asunto(s)
División Celular/genética , Senescencia Celular/genética , Epigénesis Genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/metabolismo , Animales , División Celular Asimétrica/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Agregación Celular , Linaje de la Célula/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Cromatina , Ratones Endogámicos C57BL , Transcriptoma/genética , Proteína Wnt-5a/farmacología , Proteína de Unión al GTP cdc42/metabolismo
3.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24141946

RESUMEN

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Asunto(s)
Senescencia Celular , Células Madre Hematopoyéticas/citología , Vía de Señalización Wnt , Animales , Diferenciación Celular , Polaridad Celular , Femenino , Haploinsuficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Rejuvenecimiento , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Proteína de Unión al GTP cdc42/metabolismo
5.
Aging Cell ; : e14324, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39236298

RESUMEN

Aged hematopoietic stem cells (HSCs) show reduced reconstitution potential, limiting their use in transplantation settings in the clinic. We demonstrate here that exposure of aged HSCs ex vivo to a pH of 6.9 instead of the commonly used pH of 7.4 results in enhanced HSCs potential that is consistent with rejuvenation, including attenuation of the myeloid bias of aged HSC and restoration of a youthful frequency of epigenetic polarity. Rejuvenation of aged HSCs by pH 6.9 is, at least in part, due to alterations in the polyamine/methionine pathway within pH 6.9 HSCs, and consequently, attenuation of the production of spermidine also attenuated aging of HSCs. Exposure of aged HSCs to pH 6.9, or pharmacological targeting of the polyamine pathway, might thus extend the use of HSCs from aged donors for therapeutic applications.

6.
Cell Stem Cell ; 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39270642

RESUMEN

The fundamental goal of tissue engineering is to functionally restore or improve damaged tissues or organs. Here we address this in the small bowel using an in vivo xenograft preclinical acute damage model. We investigated the therapeutic capacity of human intestinal organoids (HIOs), which are generated from human pluripotent stem cells (hPSCs), to repair damaged small bowel. We hypothesized that the HIO's cellular complexity would allow it to sustain transmural engraftment. To test this, we developed a rodent injury model where, through luminal delivery, we demonstrated that fragmented HIOs engraft, proliferate, and persist throughout the bowel following repair. Not only was restitution of the mucosal layer observed, but significant incorporation was also observed in the muscularis and vascular endothelium. Further analysis characterized sustained cell type presence within the regenerated regions, retention of proximal regionalization, and the neo-epithelia's function. These findings demonstrate the therapeutic importance of mesenchyme for intestinal injury repair.

7.
Blood Adv ; 7(11): 2590-2605, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36661340

RESUMEN

As a transcription factor in the RUNT domain core-binding factor family, RUNX1 is crucial in multiple stages of hematopoiesis, and its mutation can cause familial platelet disorder with a predisposition to acute myeloid leukemia. Previous work has established that RUNX1 is involved in the maturation of megakaryocytes (MKs) and the production of platelets. Recent studies have shown that there exists a subpopulation of hematopoietic stem cells (HSCs) with relatively high expression of von Willebrand factor and CD41 at the apex of the HSC hierarchy, termed MK-HSCs, which can give rise to MKs without going through the traditional differentiation trajectory from HSC via MPP (multipotent progenitors) and MEP (megakaryocyte-erythroid progenitor). Here, by using Runx1F/FMx1-Cre mouse model, we discovered that the MK-HSC to MK direct differentiation can occur within 1 cell division, and RUNX1 is an important regulator in the process. Runx1 knockout results in a drastic decrease in platelet counts and a severe defect in the differentiation from MK-HSCs to MKs. Single cell RNA sequencing (RNAseq) analysis shows that MK-HSCs have a distinct gene expression signature compared with non-MK-HSCs, and Runx1 deletion alters the platelet and MK-related gene expression in MK-HSCs. Furthermore, bulk RNAseq and Cut&Run analyses show that RUNX1 binds to multiple essential MK or platelet developmental genes, such as Spi1, Selp, and Itga2b and regulates their expressions in MK-HSCs. Thus, by modulating the expression of MK-related genes, RUNX1 governs the direct differentiation from MK-HSCs to MKs and platelets.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Megacariocitos , Animales , Ratones , Megacariocitos/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis , Diferenciación Celular/genética
8.
Life Sci Alliance ; 5(8)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35487692

RESUMEN

Upon aging, the function of the intestinal epithelium declines with a concomitant increase in aging-related diseases. ISCs play an important role in this process. It is known that ISC clonal dynamics follow a neutral drift model. However, it is not clear whether the drift model is still valid in aged ISCs. Tracking of clonal dynamics by clonal tracing revealed that aged crypts drift into monoclonality substantially faster than young ones. However, ISC tracing experiments, in vivo and ex vivo, implied a similar clonal expansion ability of both young and aged ISCs. Single-cell RNA sequencing for 1,920 high Lgr5 ISCs from young and aged mice revealed increased heterogeneity among subgroups of aged ISCs. Genes associated with cell adhesion were down-regulated in aged ISCs. ISCs of aged mice indeed show weaker adhesion to the matrix. Simulations applying a single cell-based model of the small intestinal crypt demonstrated an accelerated clonal drift at reduced adhesion strength, implying a central role for reduced adhesion for affecting clonal dynamics upon aging.


Asunto(s)
Intestinos , Células Madre , Animales , Células Cultivadas , Íleon , Mucosa Intestinal/metabolismo , Ratones , Células Madre/metabolismo
9.
Aging (Albany NY) ; 13(4): 4778-4793, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33629967

RESUMEN

Normal hair growth occurs in cycles, comprising growth (anagen), cessation (catagen) and rest (telogen). Upon aging, the initiation of anagen is significantly delayed, which results in impaired hair regeneration. Hair regeneration is driven by hair follicle stem cells (HFSCs). We show here that aged HFSCs present with a decrease in canonical Wnt signaling and a shift towards non-canonical Wnt5a driven signaling which antagonizes canonical Wnt signaling. Elevated expression of Wnt5a in HFSCs upon aging results in elevated activity of the small RhoGTPase Cdc42 as well as a change in the spatial distribution of Cdc42 within HFSCs. Treatment of aged HFSC with a specific pharmacological inhibitor of Cdc42 activity termed CASIN to suppress the aging-associated elevated activity of Cdc42 restored canonical Wnt signaling in aged HFSCs. Treatment of aged mice in vivo with CASIN induced anagen onset and increased the percentage of anagen skin areas. Aging-associated functional deficits of HFSCs are at least in part intrinsic to HFSCs and can be restored by rational pharmacological approaches.


Asunto(s)
Folículo Piloso/crecimiento & desarrollo , Rejuvenecimiento/fisiología , Células Madre/metabolismo , Vía de Señalización Wnt , Proteína Wnt-5a/genética , Animales , Senescencia Celular/fisiología , Ratones
10.
Leukemia ; 35(6): 1751-1762, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33077869

RESUMEN

Shwachman-Diamond syndrome (SDS) is a bone marrow failure (BMF) syndrome associated with an increased risk of myelodysplasia and leukemia. The molecular mechanisms of SDS are not fully understood. We report that primitive hematopoietic cells from SDS patients present with a reduced activity of the small RhoGTPase Cdc42 and concomitantly a reduced frequency of HSCs polar for polarity proteins. The level of apolarity of SDS HSCs correlated with the magnitude of HSC depletion in SDS patients. Importantly, exogenously provided Wnt5a or GDF11 that elevates the activity of Cdc42 restored polarity in SDS HSCs and increased the number of HSCs in SDS patient samples in surrogate ex vivo assays. Single cell level RNA-Seq analyses of SDS HSCs and daughter cells demonstrated that SDS HSC treated with GDF11 are transcriptionally more similar to control than to SDS HSCs. Treatment with GDF11 reverted pathways in SDS HSCs associated with rRNA processing and ribosome function, but also viral infection and immune function, p53-dependent DNA damage, spindle checkpoints, and metabolism, further implying a role of these pathways in HSC failure in SDS. Our data suggest that HSC failure in SDS is driven at least in part by low Cdc42 activity in SDS HSCs. Our data thus identify novel rationale approaches to attenuate HSCs failure in SDS.


Asunto(s)
Células de la Médula Ósea/citología , Polaridad Celular , Células Madre Hematopoyéticas/citología , Síndrome de Shwachman-Diamond/prevención & control , Proteína de Unión al GTP cdc42/metabolismo , Células de la Médula Ósea/metabolismo , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Pronóstico , Síndrome de Shwachman-Diamond/etiología , Síndrome de Shwachman-Diamond/metabolismo , Síndrome de Shwachman-Diamond/patología , Proteína Wnt-5a/química , Proteína Wnt-5a/metabolismo , Proteína de Unión al GTP cdc42/química
11.
J Clin Invest ; 117(11): 3283-95, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17960249

RESUMEN

The molecular pathogenesis of the myeloid leukemias that frequently occur in patients with Fanconi anemia (FA) is not well defined. Hematopoietic stem cells bearing inactivating mutations of FA complementation group C (FANCC) are genetically unstable and hypersensitive to apoptotic cytokine cues including IFN-gamma and TNF-alpha, but neoplastic stem cell clones that arise frequently in vivo are resistant to these cytokines. Reasoning that the combination of genetic instability and cytokine hypersensitivity might create an environment supporting the emergence of leukemic stem cells, we tested the leukemia-promoting effects of TNF-alpha in murine stem cells. TNF-alpha exposure initially profoundly inhibited the growth of Fancc-/- stem cells. However, longer-term exposure of these cells promoted the outgrowth of cytogenetically abnormal clones that, upon transplantation into congenic WT mice, led to acute myelogenous leukemia. TNF-alpha induced ROS-dependent genetic instability in Fancc-/- but not in WT cells. The leukemic clones were TNF-alpha resistant but retained their characteristic hypersensitivity to mitomycin C and exhibited high levels of chromosomal instability. Expression of FANCC cDNA in Fancc-/- stem cells protected them from TNF-alpha-induced clonal evolution. We conclude that TNF-alpha exposure creates an environment in which somatically mutated preleukemic stem cell clones are selected and from which unaltered TNF-alpha-hypersensitive Fancc-/- stem cells are purged.


Asunto(s)
Proliferación Celular , Proteína del Grupo de Complementación C de la Anemia de Fanconi/inmunología , Anemia de Fanconi/inmunología , Células Madre Hematopoyéticas/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Apoptosis , Diferenciación Celular/fisiología , Aberraciones Cromosómicas , Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Prueba de Complementación Genética , Células Madre Hematopoyéticas/citología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trasplante de Células Madre , Tasa de Supervivencia
12.
Blood Adv ; 4(21): 5512-5526, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33166407

RESUMEN

Hematopoietic stem cell (HSC) activity is tightly controlled to ensure the integrity of the hematopoietic system during the organism's lifetime. How the HSC compartment maintains its long-term fitness in conditions of chronic stresses associated with systemic metabolic disorders is poorly understood. In this study, we show that obesity functionally affects the long-term function of the most immature engrafting HSC subpopulation. We link this altered regenerative activity to the oxidative stress and the aberrant constitutive activation of the AKT signaling pathway that characterized the obese environment. In contrast, we found minor disruptions of the HSC function in obese mice at steady state, suggesting that active mechanisms could protect the HSC compartment from its disturbed environment. Consistent with this idea, we found that FOXO proteins in HSCs isolated from obese mice become insensitive to their normal upstream regulators such as AKT, even during intense oxidative stress. We established that hyperglycemia, a key condition associated with obesity, is directly responsible for the alteration of the AKT-FOXO axis in HSCs and their abnormal oxidative stress response. As a consequence, we observed that HSCs isolated from a hyperglycemic environment display enhanced resistance to oxidative stress and DNA damage. Altogether, these results indicate that chronic metabolic stresses associated with obesity and/or hyperglycemia affect the wiring of the HSCs and modify their oxidative stress response. These data suggest that the uncoupling of FOXO from its environmental regulators could be a key adaptive strategy that promotes the survival of the HSC compartment in obesity.


Asunto(s)
Células Madre Hematopoyéticas , Hiperglucemia , Animales , Daño del ADN , Ratones , Estrés Oxidativo , Transducción de Señal
13.
Aging Cell ; 19(9): e13208, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32755011

RESUMEN

Cdc42 is a small RhoGTPase regulating multiple functions in eukaryotic cells. The activity of Cdc42 is significantly elevated in several tissues of aged mice, while the Cdc42 gain-of-activity mouse model presents with a premature aging-like phenotype and with decreased lifespan. These data suggest a causal connection between elevated activity of Cdc42, aging, and reduced lifespan. Here, we demonstrate that systemic treatment of aged (75-week-old) female C57BL/6 mice with a Cdc42 activity-specific inhibitor (CASIN) for 4 consecutive days significantly extends average and maximum lifespan. Moreover, aged CASIN-treated animals displayed a youthful level of the aging-associated cytokines IL-1ß, IL-1α, and INFγ in serum and a significantly younger epigenetic clock as based on DNA methylation levels in blood cells. Overall, our data show that systemic administration of CASIN to reduce Cdc42 activity in aged mice extends murine lifespan.


Asunto(s)
Citocinas/metabolismo , Proteína de Unión al GTP cdc42/genética , Envejecimiento , Animales , Proteínas de Drosophila , Femenino , Cadenas alfa de Integrinas , Longevidad , Ratones , Ratones Endogámicos C57BL
14.
Methods Mol Biol ; 506: 423-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19110642

RESUMEN

Maintaining the stability of the genome is critical to cell survival and normal cell growth. Genetic modification of hematopoietic cells might bear an inherent increased risk for the accumulation of DNA mutations. It frequently requires cultivation of the cells under super-physiological oxygen levels, which can result in increased oxidative damage, as well as under super-physiological concentrations of cytokines, which might interfere with DNA-damage checkpoint activation and by this means might result in an increased mutational load. We describe here a protocol for monitoring the frequency of DNA mutations in bone marrow cells post transduction or upon selection either in vitro or in vivo based on the lacZ-plasmid (pUR288) transgenic mouse (small blue mouse) mutation indicator strain.


Asunto(s)
Genómica , Células Madre Hematopoyéticas/metabolismo , Mutación , Animales , Secuencia de Bases , Cartilla de ADN , Electroporación , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
15.
Exp Hematol ; 59: 40-50.e3, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29288703

RESUMEN

Self-renewing hematopoietic stem cells and multipotent progenitor cells are responsible for maintaining hematopoiesis throughout an individual's lifetime. For overall health and survival, it is critical that the genome stability of these cells is maintained and that the cell population is not exhausted. Previous reports have indicated that the DEK protein, a chromatin structural protein that functions in numerous nuclear processes, is required for DNA damage repair in vitro and long-term engraftment of hematopoietic stem cells in vivo. Therefore, we investigated the role of DEK in normal hematopoiesis and response to DNA damaging agents in vivo. Here, we report that hematopoiesis is largely unperturbed in DEK knockout mice compared with wild-type (WT) controls. However, DEK knockout mice have fewer radioprotective units, but increased capacity to survive repeated sublethal doses of radiation exposure compared with WT mice. Furthermore, this increased survival correlated with a sustained quiescent state in which DEK knockout restricted hematopoietic progenitor cells (HPC-1) were nearly three times more likely to be quiescent following irradiation compared with WT cells and were significantly more radioresistant during the early phases of myeloid reconstitution. Together, our studies indicate that DEK functions in the normal hematopoietic stress response to recurrent radiation exposure.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/deficiencia , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Oncogénicas/deficiencia , Proteínas de Unión a Poli-ADP-Ribosa/deficiencia , Tolerancia a Radiación/fisiología , Animales , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados
16.
Stem Cell Reports ; 9(5): 1359-1368, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29056333

RESUMEN

The spindle assembly checkpoint plays a pivotal role in preventing aneuploidy and transformation. Many studies demonstrate impairment of this checkpoint in cancer cells. While leukemia is frequently driven by transformed hematopoietic stem and progenitor cells (HSPCs), the biology of the spindle assembly checkpoint in such primary cells is not very well understood. Here, we reveal that the checkpoint is fully functional in murine progenitor cells and, to a lesser extent, in hematopoietic stem cells. We show that HSPCs arrest at prometaphase and induce p53-dependent apoptosis upon prolonged treatment with anti-mitotic drugs. Moreover, the checkpoint can be chemically and genetically abrogated, leading to premature exit from mitosis, subsequent enforced G1 arrest, and enhanced levels of chromosomal damage. We finally demonstrate that, upon checkpoint abrogation in HSPCs, hematopoiesis is impaired, manifested by loss of differentiation potential and engraftment ability, indicating a critical role of this checkpoint in HSPCs and hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Puntos de Control de la Fase M del Ciclo Celular , Animales , Antimitóticos/farmacología , Apoptosis , Células Cultivadas , Hematopoyesis , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Cell Rep ; 18(11): 2608-2621, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28297666

RESUMEN

Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.


Asunto(s)
Senescencia Celular , Intestino Delgado/citología , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt , Animales , Biomarcadores/metabolismo , Recuento de Células , Proliferación Celular , Femenino , Ratones , Organoides/citología , Regeneración , Nicho de Células Madre
18.
Cell Rep ; 13(11): 2412-2424, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26686632

RESUMEN

Whether aged hematopoietic stem and progenitor cells (HSPCs) have impaired DNA damage repair is controversial. Using a combination of DNA mutation indicator assays, we observe a 2- to 3-fold increase in the number of DNA mutations in the hematopoietic system upon aging. Young and aged hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) do not show an increase in mutation upon irradiation-induced DNA damage repair, and young and aged HSPCs respond very similarly to DNA damage with respect to cell-cycle checkpoint activation and apoptosis. Both young and aged HSPCs show impaired activation of the DNA-damage-induced G1-S checkpoint. Induction of chronic DNA double-strand breaks by zinc-finger nucleases suggests that HSPCs undergo apoptosis rather than faulty repair. These data reveal a protective mechanism in both the young and aged hematopoietic system against accumulation of mutations in response to DNA damage.


Asunto(s)
Envejecimiento , Genoma , Células Madre Hematopoyéticas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de la radiación , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Células Cultivadas , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Daño del ADN/efectos de la radiación , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de la radiación , Rayos gamma , Células Madre Hematopoyéticas/citología , Pérdida de Heterocigocidad , Ratones , Ratones Endogámicos C57BL , Mutación , Puntos de Control de la Fase S del Ciclo Celular/efectos de la radiación , Trasplante Homólogo , Irradiación Corporal Total
19.
Nat Med ; 18(7): 1123-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22729286

RESUMEN

Tissue damage induced by ionizing radiation in the hematopoietic and gastrointestinal systems is the major cause of lethality in radiological emergency scenarios and underlies some deleterious side effects in patients undergoing radiation therapy. The identification of target-specific interventions that confer radiomitigating activity is an unmet challenge. Here we identify the thrombomodulin (Thbd)-activated protein C (aPC) pathway as a new mechanism for the mitigation of total body irradiation (TBI)-induced mortality. Although the effects of the endogenous Thbd-aPC pathway were largely confined to the local microenvironment of Thbd-expressing cells, systemic administration of soluble Thbd or aPC could reproduce and augment the radioprotective effect of the endogenous Thbd-aPC pathway. Therapeutic administration of recombinant, soluble Thbd or aPC to lethally irradiated wild-type mice resulted in an accelerated recovery of hematopoietic progenitor activity in bone marrow and a mitigation of lethal TBI. Starting infusion of aPC as late as 24 h after exposure to radiation was sufficient to mitigate radiation-induced mortality in these mice. These findings suggest that pharmacologic augmentation of the activity of the Thbd-aPC pathway by recombinant Thbd or aPC might offer a rational approach to the mitigation of tissue injury and lethality caused by ionizing radiation.


Asunto(s)
Proteína C/antagonistas & inhibidores , Traumatismos por Radiación/prevención & control , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Trombomodulina/antagonistas & inhibidores , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Receptores de Trombina , Análisis de Supervivencia , Trombomodulina/genética , Trombomodulina/metabolismo , Irradiación Corporal Total
20.
Nat Med ; 16(10): 1141-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20871610

RESUMEN

Mobilization of hematopoietic stem and progenitor cells (HSPCs) from bone marrow into peripheral blood by the cytokine granulocyte colony-stimulating factor (G-CSF) has become the preferred source of HSPCs for stem cell transplants. However, G-CSF fails to mobilize sufficient numbers of stem cells in up to 10% of donors, precluding autologous transplantation in those donors or substantially delaying transplant recovery time. Consequently, new regimens are needed to increase the number of stem cells in peripheral blood upon mobilization. Using a forward genetic approach in mice, we mapped the gene encoding the epidermal growth factor receptor (Egfr) to a genetic region modifying G-CSF-mediated HSPC mobilization. Amounts of EGFR in HSPCs inversely correlated with the cells' ability to be mobilized by G-CSF, implying a negative role for EGFR signaling in mobilization. In combination with G-CSF treatment, genetic reduction of EGFR activity in HSPCs (in waved-2 mutant mice) or treatment with the EGFR inhibitor erlotinib increased mobilization. Increased mobilization due to suppression of EGFR activity correlated with reduced activity of cell division control protein-42 (Cdc42), and genetic Cdc42 deficiency in vivo also enhanced G-CSF-induced mobilization. Our findings reveal a previously unknown signaling pathway regulating stem cell mobilization and provide a new pharmacological approach for improving HSPC mobilization and thereby transplantation outcomes.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Transducción de Señal , Animales , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteína de Unión al GTP cdc42/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA