Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(10): 1060-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26343536

RESUMEN

Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a(-/-) LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a(-/-) LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células de Langerhans/efectos de la radiación , Radiación Ionizante , Linfocitos T Reguladores/efectos de la radiación , Animales , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Citometría de Flujo , Ratones , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Regulación hacia Arriba
2.
Br J Haematol ; 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38946206

RESUMEN

Erythroid cells undergo a highly complex maturation process, resulting in dynamic changes that generate red blood cells (RBCs) highly rich in haemoglobin. The end stages of the erythroid cell maturation process primarily include chromatin condensation and nuclear polarization, followed by nuclear expulsion called enucleation and clearance of mitochondria and other organelles to finally generate mature RBCs. While healthy RBCs are devoid of mitochondria, recent evidence suggests that mitochondria are actively implicated in the processes of erythroid cell maturation, erythroblast enucleation and RBC production. However, the extent of mitochondrial participation that occurs during these ultimate steps is not completely understood. This is specifically important since abnormal RBC retention of mitochondria or mitochondrial DNA contributes to the pathophysiology of sickle cell and other disorders. Here we review some of the key findings so far that elucidate the importance of this process in various aspects of erythroid maturation and RBC production under homeostasis and disease conditions.

3.
Curr Opin Hematol ; 30(1): 1-3, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36473018

RESUMEN

PURPOSE OF REVIEW: Quiescence is a fundamental property of haematopoietic stem cells (HSCs). Despite the importance of quiescence in predicting the potency of HSCs, tools that measure routinely the degree of quiescence or select for quiescent HSCs have been lacking. This Commentary discusses recent findings that address this fundamental gap in the HSC toolbox. RECENT FINDINGS: Highly purified, phenotypically-defined HSCs are heterogeneous in their mitochondrial membrane potential (MMP). The lowest MMP subsets are enriched in greatly quiescent HSCs with the highest potency within the purified HSC population. MMP provides an intrinsic probe to select HSC subsets with unique cell cycle properties and distinct stem cell potential. Using this approach, new and unanticipated metabolic properties of quiescent HSCs' exit have been discovered. This methodology may improve the mechanistic understanding, of HSCs' exit from and entry to, quiescence. SUMMARY: Selecting HSCs using MMP is likely to lead to discoveries of new HSC properties, may improve the ex vivo maintenance of HSCs and has implications for the clinic, including for improving HSC transplantations.


Asunto(s)
Células Madre Hematopoyéticas , Humanos , Potencial de la Membrana Mitocondrial
4.
Blood ; 133(18): 1943-1952, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30808633

RESUMEN

The hematopoietic system produces new blood cells throughout life. Mature blood cells all derived from a pool of rare long-lived hematopoietic stem cells (HSCs) that are mostly quiescent but occasionally divide and self-renew to maintain the stem cell pool and to insure the continuous replenishment of blood cells. Mitochondria have recently emerged as critical not only for HSC differentiation and commitment but also for HSC homeostasis. Mitochondria are dynamic organelles that orchestrate a number of fundamental metabolic and signaling processes, producing most of the cellular energy via oxidative phosphorylation. HSCs have a relatively high amount of mitochondria that are mostly inactive. Here, we review recent advances in our understanding of the role of mitochondria in HSC homeostasis and discuss, among other topics, how mitochondrial dynamism and quality control might be implicated in HSC fate, self-renewal, and regenerative potential.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Mitocondrias/metabolismo , Animales , Células Madre Hematopoyéticas/citología , Homeostasis/fisiología , Humanos
5.
Am J Hematol ; 94(1): 10-20, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30252956

RESUMEN

The signaling cascade induced by the interaction of erythropoietin (EPO) with its receptor (EPO-R) is a key event of erythropoiesis. We present here data indicating that Fyn, a Src-family-kinase, participates in the EPO signaling-pathway, since Fyn-/- mice exhibit reduced Tyr-phosphorylation of EPO-R and decreased STAT5-activity. The importance of Fyn in erythropoiesis is also supported by the blunted responsiveness of Fyn-/- mice to stress erythropoiesis. Fyn-/- mouse erythroblasts adapt to reactive oxygen species (ROS) by activating the redox-related-transcription-factor Nrf2. However, since Fyn is a physiologic repressor of Nrf2, absence of Fyn resulted in persistent-activation of Nrf2 and accumulation of nonfunctional proteins. ROS-induced over-activation of Jak2-Akt-mTOR-pathway and repression of autophagy with perturbation of lysosomal-clearance were also noted. Treatment with Rapamycin, a mTOR-inhibitor and autophagy activator, ameliorates Fyn-/- mouse baseline erythropoiesis and erythropoietic response to oxidative-stress. These findings identify a novel multimodal action of Fyn in the regulation of normal and stress erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-fyn/fisiología , Animales , Autofagia , Doxorrubicina/toxicidad , Eritroblastos/enzimología , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Femenino , Janus Quinasa 2/metabolismo , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Fenilhidrazinas/toxicidad , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-fyn/deficiencia , Proteínas Proto-Oncogénicas c-fyn/genética , Especies Reactivas de Oxígeno , Receptores de Eritropoyetina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
6.
Curr Opin Hematol ; 25(4): 290-298, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29782339

RESUMEN

PURPOSE OF REVIEW: Work in the past decade has revealed key functions of the evolutionary conserved transcription factors Forkhead box O (FOXO) in the maintenance of homeostatic hematopoiesis. Here the diverse array of FOXO functions in normal and diseased hematopoietic stem and progenitor cells is reviewed and the main findings in the past decade are highlighted. Future work should reveal FOXO-regulated networks whose alterations contribute to hematological disorders. RECENT FINDINGS: Recent studies have identified unanticipated FOXO functions in hematopoiesis including in hematopoietic stem and progenitor cells (HSPC), erythroid cells, and immune cells. These findings suggest FOXO3 is critical for the regulation of mitochondrial and metabolic processes in hematopoietic stem cells, the balanced lineage determination, the T and B homeostasis, and terminal erythroblast maturation and red blood cell production. In aggregate these findings highlight the context-dependent function of FOXO in hematopoietic cells. Recent findings also question the nature of FOXO's contribution to heme malignancies as well as the mechanisms underlying FOXO's regulation in HSPC. SUMMARY: FOXO are safeguards of homeostatic hematopoiesis. FOXO networks and their regulators and coactivators in HSPC are greatly complex and less well described. Identifications and characterizations of these FOXO networks in disease are likely to uncover disease-promoting mechanisms.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Enfermedades Hematológicas/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Animales , Enfermedades Hematológicas/patología , Células Madre Hematopoyéticas/patología , Humanos
7.
J Biol Chem ; 292(7): 3005-3015, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-27994057

RESUMEN

Accumulation of damaged DNA in hematopoietic stem cells (HSC) is associated with chromosomal abnormalities, genomic instability, and HSC aging and might promote hematological malignancies with age. Despite this, the regulatory pathways implicated in the HSC DNA damage response have not been fully elucidated. One of the sources of DNA damage is reactive oxygen species (ROS) generated by both exogenous and endogenous insults. Balancing ROS levels in HSC requires FOXO3, which is an essential transcription factor for HSC maintenance implicated in HSC aging. Elevated ROS levels result in defective Foxo3-/- HSC cycling, among many other deficiencies. Here, we show that loss of FOXO3 leads to the accumulation of DNA damage in primitive hematopoietic stem and progenitor cells (HSPC), associated specifically with reduced expression of genes implicated in the repair of oxidative DNA damage. We provide further evidence that Foxo3-/- HSPC are defective in DNA damage repair. Specifically, we show that the base excision repair pathway, the main pathway utilized for the repair of oxidative DNA damage, is compromised in Foxo3-/- primitive hematopoietic cells. Treating mice in vivo with N-acetylcysteine reduces ROS levels, rescues HSC cycling defects, and partially mitigates HSPC DNA damage. These results indicate that DNA damage accrued as a result of elevated ROS in Foxo3-/- mutant HSPC is at least partially reversible. Collectively, our findings suggest that FOXO3 serves as a protector of HSC genomic stability and health.


Asunto(s)
Daño del ADN , Proteína Forkhead Box O3/fisiología , Células Madre Hematopoyéticas/citología , Estrés Oxidativo , Acetilcisteína/farmacología , Animales , Ciclo Celular/fisiología , Proteína Forkhead Box O3/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
8.
EMBO J ; 33(2): 93-5, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24421322

RESUMEN

Epigenetic modifications of stem cell genome including DNA methylation and histone modifications are critical for the regulation of stem cell gene expression and maintenance of stem cell pool and their differentiation. Although the importance of epigenetic modifications specifically DNA methylation to adult hematopoietic stem cells (HSC) has been established, the identity of specific modulators and precise mechanism of integration of methylation events remain to be uncovered. In this issue, Shuai and colleagues identify the SUMO E3 ligase PIAS1 (protein inhibitor of activated STAT1) as a key regulator of DNA methylation of HSC required for their maintenance and lineage commitment (Liu et al, 2014).


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , Células Madre Hematopoyéticas/fisiología , Proteínas Inhibidoras de STAT Activados/fisiología , Animales
9.
PLoS Genet ; 11(10): e1005526, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26452208

RESUMEN

Circulating red blood cells (RBCs) are essential for tissue oxygenation and homeostasis. Defective terminal erythropoiesis contributes to decreased generation of RBCs in many disorders. Specifically, ineffective nuclear expulsion (enucleation) during terminal maturation is an obstacle to therapeutic RBC production in vitro. To obtain mechanistic insights into terminal erythropoiesis we focused on FOXO3, a transcription factor implicated in erythroid disorders. Using an integrated computational and experimental systems biology approach, we show that FOXO3 is essential for the correct temporal gene expression during terminal erythropoiesis. We demonstrate that the FOXO3-dependent genetic network has critical physiological functions at key steps of terminal erythropoiesis including enucleation and mitochondrial clearance processes. FOXO3 loss deregulated transcription of genes implicated in cell polarity, nucleosome assembly and DNA packaging-related processes and compromised erythroid enucleation. Using high-resolution confocal microscopy and imaging flow cytometry we show that cell polarization is impaired leading to multilobulated Foxo3-/- erythroblasts defective in nuclear expulsion. Ectopic FOXO3 expression rescued Foxo3-/- erythroblast enucleation-related gene transcription, enucleation defects and terminal maturation. Remarkably, FOXO3 ectopic expression increased wild type erythroblast maturation and enucleation suggesting that enhancing FOXO3 activity may improve RBCs production. Altogether these studies uncover FOXO3 as a novel regulator of erythroblast enucleation and terminal maturation suggesting FOXO3 modulation might be therapeutic in disorders with defective erythroid maturation.


Asunto(s)
Eritrocitos/metabolismo , Eritropoyesis/genética , Factores de Transcripción Forkhead/genética , Biología de Sistemas , Animales , Autofagia/genética , Células de la Médula Ósea/metabolismo , Polaridad Celular/genética , Eritroblastos/metabolismo , Eritrocitos/citología , Citometría de Flujo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Homeostasis , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo
10.
Genes Dev ; 24(15): 1620-33, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20679398

RESUMEN

The bicistronic microRNA (miRNA) locus miR-144/451 is highly expressed during erythrocyte development, although its physiological roles are poorly understood. We show that miR-144/451 ablation in mice causes mild erythrocyte instability and increased susceptibility to damage after exposure to oxidant drugs. This phenotype is deeply conserved, as miR-451 depletion synergizes with oxidant stress to cause profound anemia in zebrafish embryos. At least some protective activities of miR-451 stem from its ability to directly suppress production of 14-3-3zeta, a phospho-serine/threonine-binding protein that inhibits nuclear accumulation of transcription factor FoxO3, a positive regulator of erythroid anti-oxidant genes. Thus, in miR-144/451(-/-) erythroblasts, 14-3-3zeta accumulates, causing partial relocalization of FoxO3 from nucleus to cytoplasm with dampening of its transcriptional program, including anti-oxidant-encoding genes Cat and Gpx1. Supporting this mechanism, overexpression of 14-3-3zeta in erythroid cells and fibroblasts inhibits nuclear localization and activity of FoxO3. Moreover, shRNA suppression of 14-3-3zeta protects miR-144/451(-/-) erythrocytes against peroxide-induced destruction, and restores catalase activity. Our findings define a novel miRNA-regulated pathway that protects erythrocytes against oxidant stress, and, more generally, illustrate how a miRNA can influence gene expression by altering the activity of a key transcription factor.


Asunto(s)
Proteínas 14-3-3/metabolismo , Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Estrés Oxidativo , Proteínas 14-3-3/genética , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Catalasa/metabolismo , Células Eritroides/enzimología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , MicroARNs/genética , Alineación de Secuencia , Eliminación de Secuencia/genética , Pez Cebra/genética , Pez Cebra/metabolismo
11.
Development ; 141(22): 4206-18, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25371358

RESUMEN

An appropriate balance between self-renewal and differentiation is crucial for stem cell function during both early development and tissue homeostasis throughout life. Recent evidence from both pluripotent embryonic and adult stem cell studies suggests that this balance is partly regulated by reactive oxygen species (ROS), which, in synchrony with metabolism, mediate the cellular redox state. In this Primer, we summarize what ROS are and how they are generated in the cell, as well as their downstream molecular targets. We then review recent findings that provide molecular insights into how ROS signaling can influence stem cell homeostasis and lineage commitment, and discuss the implications of this for reprogramming and stem cell ageing. We conclude that ROS signaling is an emerging key regulator of multiple stem cell populations.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Homeostasis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Humanos , Oxidación-Reducción
12.
J Bioenerg Biomembr ; 49(4): 343-346, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28639090

RESUMEN

The production of all blood cells from hematopoietic stem cells (HSC) is highly sensitive to reactive oxygen species (ROS). Cumulating evidence suggests that mitochondria are critical for HSC fate determination. FOXO are known regulators of anti-oxidant response and key to the maintenance of HSC. Recent works indicate that FOXO3 is implicated in the control of mitochondrial function beyond regulating levels of ROS in HSC. Here we review these findings and discuss implications for homeostatic blood formation and stem cell fate determination.


Asunto(s)
Linaje de la Célula , Proteína Forkhead Box O3/fisiología , Células Madre Hematopoyéticas/fisiología , Homeostasis , Animales , Células Sanguíneas/citología , Humanos , Mitocondrias/metabolismo , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo
13.
EMBO Rep ; 16(9): 1164-76, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26209246

RESUMEN

Hematopoietic stem cells (HSC) are primarily dormant but have the potential to become highly active on demand to reconstitute blood. This requires a swift metabolic switch from glycolysis to mitochondrial oxidative phosphorylation. Maintenance of low levels of reactive oxygen species (ROS), a by-product of mitochondrial metabolism, is also necessary for sustaining HSC dormancy. Little is known about mechanisms that integrate energy metabolism with hematopoietic stem cell homeostasis. Here, we identify the transcription factor FOXO3 as a new regulator of metabolic adaptation of HSC. ROS are elevated in Foxo3(-/-) HSC that are defective in their activity. We show that Foxo3(-/-) HSC are impaired in mitochondrial metabolism independent of ROS levels. These defects are associated with altered expression of mitochondrial/metabolic genes in Foxo3(-/-) hematopoietic stem and progenitor cells (HSPC). We further show that defects of Foxo3(-/-) HSC long-term repopulation activity are independent of ROS or mTOR signaling. Our results point to FOXO3 as a potential node that couples mitochondrial metabolism with HSC homeostasis. These findings have critical implications for mechanisms that promote malignant transformation and aging of blood stem and progenitor cells.


Asunto(s)
Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Envejecimiento/genética , Animales , Proteína Forkhead Box O3 , Homeostasis/genética , Homeostasis/fisiología , Ratones , Mitocondrias/genética , Estrés Oxidativo , Serina-Treonina Quinasas TOR/metabolismo
14.
Br J Haematol ; 174(5): 661-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27442953

RESUMEN

Anaemia or decreased blood haemoglobin is the most common blood disorder often characterized by reduced red blood cell (RBC) numbers. RBCs are produced from differentiation and commitment of haematopoietic stem cells to the erythroid lineage by a process called erythropoiesis. Coordination of erythropoietin receptor signalling with several erythroid transcription factors including GATA1 is essential for this process. A number of additional players that are critical for RBC production have been identified in recent years. Major technological advances, such as the development of RNA interference, genetically modified animals, including zebrafish, and imaging flow cytometry have led to these discoveries; the emergence of -omics approaches in combination with the optimization of ex vivo erythroid cultures have also produced a more comprehensive understanding of erythropoiesis. Here we summarize studies describing novel regulators of erythropoiesis that modulate erythroid cell production in the context of human erythroid disorders involving hypoxia, iron regulation, immune-related molecules, and the transcription factor FOXO3.


Asunto(s)
Células Eritroides/patología , Eritropoyesis/fisiología , Homeostasis/fisiología , Animales , Proteína Forkhead Box O3/metabolismo , Humanos , Hipoxia , Enfermedades del Sistema Inmune/patología , Hierro/metabolismo
16.
Blood ; 124(14): 2285-97, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25115889

RESUMEN

Complex genetic networks control hematopoietic stem cell differentiation into progenitors that give rise to billions of erythrocytes daily. Previously, we described a role for the master regulator of erythropoiesis, GATA-1, in inducing genes encoding components of the autophagy machinery. In this context, the Forkhead transcription factor, Foxo3, amplified GATA-1-mediated transcriptional activation. To determine the scope of the GATA-1/Foxo3 cooperativity, and to develop functional insights, we analyzed the GATA-1/Foxo3-dependent transcriptome in erythroid cells. GATA-1/Foxo3 repressed expression of Exosc8, a pivotal component of the exosome complex, which mediates RNA surveillance and epigenetic regulation. Strikingly, downregulating Exosc8, or additional exosome complex components, in primary erythroid precursor cells induced erythroid cell maturation. Our results demonstrate a new mode of controlling erythropoiesis in which multiple components of the exosome complex are endogenous suppressors of the erythroid developmental program.


Asunto(s)
Eritrocitos/citología , Exosomas/fisiología , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA1/metabolismo , Animales , Autofagia , Diferenciación Celular , Epigénesis Genética , Eritroblastos/citología , Células Eritroides/metabolismo , Eritropoyesis/genética , Proteína Forkhead Box O3 , Regulación de la Expresión Génica , Ratones , ARN/metabolismo , Activación Transcripcional
17.
EMBO J ; 29(24): 4118-31, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21113129

RESUMEN

Reactive oxygen species (ROS) participate in normal intracellular signalling and in many diseases including cancer and aging, although the associated mechanisms are not fully understood. Forkhead Box O (FoxO) 3 transcription factor regulates levels of ROS concentrations, and is essential for maintenance of hematopoietic stem cells. Here, we show that loss of Foxo3 causes a myeloproliferative syndrome with splenomegaly and increased hematopoietic progenitors (HPs) that are hypersensitive to cytokines. These mutant HPs contain increased ROS, overactive intracellular signalling through the AKT/mammalian target of rapamycin signalling pathway and relative deficiency of Lnk, a negative regulator of cytokine receptor signalling. In vivo treatment with ROS scavenger N-acetyl-cysteine corrects these biochemical abnormalities and relieves the myeloproliferation. Moreover, enforced expression of Lnk by retroviral transfer corrects the abnormal expansion of Foxo3(-/-) HPs in vivo. Our combined results show that loss of Foxo3 causes increased ROS accumulation in HPs. In turn, this inhibits Lnk expression that contributes to exaggerated cytokine responses that lead to myeloproliferation. Our findings could explain the mechanisms by which mutations that alter Foxo3 function induce malignancy. More generally, the work illustrates how deregulated ROS may contribute to malignant progression.


Asunto(s)
Factores de Transcripción Forkhead/deficiencia , Trastornos Mieloproliferativos/patología , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Acetilcisteína/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteína Forkhead Box O3 , Depuradores de Radicales Libres/uso terapéutico , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones Noqueados , Trastornos Mieloproliferativos/tratamiento farmacológico , Transducción de Señal , Esplenomegalia/patología
18.
Haematologica ; 99(2): 267-75, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23975182

RESUMEN

Resveratrol, a polyphenolic-stilbene, has received increased attention in the last decade due to its wide range of biological activities. Beta(ß)-thalassemias are inherited red cell disorders, found worldwide, characterized by ineffective erythropoiesis and red cell oxidative damage with reduced survival. We evaluated the effects of low-dose-resveratrol (5 µM) on in vitro human erythroid differentiation of CD34(+) from normal and ß-thalassemic subjects. We found that resveratrol induces accelerated erythroid-maturation, resulting in the reduction of colony-forming units of erythroid cells and increased intermediate and late erythroblasts. In sorted colony-forming units of erythroid cells resveratrol activates Forkhead-box-class-O3, decreases Akt activity and up-regulates anti-oxidant enzymes as catalase. In an in vivo murine model for ß-thalassemia, resveratrol (2.4 mg/kg) reduces ineffective erythropoiesis, increases hemoglobin levels, reduces reticulocyte count and ameliorates red cell survival. In both wild-type and ß-thalassemic mice, resveratrol up-regulates scavenging enzymes such as catalase and peroxiredoxin-2 through Forkhead-box-class-O3 activation. These data indicate that resveratrol inhibits Akt resulting in FoxO3 activation with upregulation of cytoprotective systems enabling the pathological erythroid precursors to resist the oxidative damage and continue to differentiate. Our data suggest that the dual effect of resveratrol on erythropoiesis through activation of FoxO3 transcriptional factor combined with the amelioration of oxidative stress in circulating red cells may be considered as a potential novel therapeutic strategy in treating ß-thalassemia.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Eritrocitos/metabolismo , Eritropoyesis/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Estilbenos/farmacología , Talasemia beta/metabolismo , Animales , Catalasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritrocitos/patología , Proteína Forkhead Box O3 , Humanos , Masculino , Ratones , Peroxirredoxinas/metabolismo , Resveratrol , Talasemia beta/tratamiento farmacológico , Talasemia beta/patología
19.
Am J Hematol ; 89(12): E235-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25163926

RESUMEN

Resveratrol is a plant-derived polyphenol that has shown protective effects against many disorders including, several types of cancers and other age-associated diseases as well as blood disorders in cultured cells and/or animal models. However, whether resveratrol has any impact specifically on normal blood stem cells remains unknown. Here, we show that a 3-week treatment of resveratrol increases the frequency and total numbers of normal bone marrow hematopoietic stem cells (HSC) without any impact on their competitive repopulation capacity. In addition, we show that resveratrol enhances the bone marrow multipotent progenitor capacity in vivo. These results have therapeutic value for disorders of hematopoietic stem and progenitor cells (HSPC) as well as for bone marrow transplantation settings.


Asunto(s)
Antioxidantes/farmacología , Células de la Médula Ósea/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Rayos gamma , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inyecciones Intraperitoneales , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Resveratrol , Estilbenos , Irradiación Corporal Total
20.
Am J Hematol ; 89(10): 954-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24966026

RESUMEN

Ineffective erythropoiesis is observed in many erythroid disorders including ß-thalassemia and anemia of chronic disease in which increased production of erythroblasts that fail to mature exacerbate the underlying anemias. As loss of the transcription factor FOXO3 results in erythroblast abnormalities similar to the ones observed in ineffective erythropoiesis, we investigated the underlying mechanisms of the defective Foxo3(-/-) erythroblast cell cycle and maturation. Here we show that loss of Foxo3 results in overactivation of the JAK2/AKT/mTOR signaling pathway in primary bone marrow erythroblasts partly mediated by redox modulation. We further show that hyperactivation of mTOR signaling interferes with cell cycle progression in Foxo3 mutant erythroblasts. Importantly, inhibition of mTOR signaling, in vivo or in vitro enhances significantly Foxo3 mutant erythroid cell maturation. Similarly, in vivo inhibition of mTOR remarkably improves erythroid cell maturation and anemia in a model of ß-thalassemia. Finally we show that FOXO3 and mTOR are likely part of a larger metabolic network in erythroblasts as together they control the expression of an array of metabolic genes some of which are implicated in erythroid disorders. These combined findings indicate that a metabolism-mediated regulatory network centered by FOXO3 and mTOR control the balanced production and maturation of erythroid cells. They also highlight physiological interactions between these proteins in regulating erythroblast energy. Our results indicate that alteration in the function of this network might be implicated in the pathogenesis of ineffective erythropoiesis.


Asunto(s)
Eritroblastos/metabolismo , Eritropoyesis , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Modelos Animales de Enfermedad , Eritroblastos/patología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Ratones , Ratones Noqueados , Serina-Treonina Quinasas TOR/genética , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA