Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
2.
PLoS One ; 18(10): e0293027, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37844090

RESUMEN

A potential direct correlation between systemic inflammation and physiological aging has been suggested, along with whether there is a higher expression of inflammatory markers in otherwise healthy older adults. Cross-sectional data were extracted from the publicly available 2016 Health and Retirement Survey, a nationally representative survey of older adults in the United States. A subset of participants (n = 9934) consented to a blood draw at the time of recruitment and were measured for high sensitivity C-reactive protein (hs-CRP), Interleukin (IL-6, IL-10, IL-1RA), soluble tumor necrosis factor receptor (sTNFR-1) and transforming growth factor beta 1 (TGF-ß1). We included 9,188 participants, representative of 83,939,225 nationally. After adjusting for sex and the number of comorbidities, there remained a significant positive correlation between age and ln (log adjusted) IL-6, and ln sTNFR-1, and a significant inverse correlation between age and ln IL-1RA, ln TGF-ß1, and ln hs-CRP. Among the subset of participants who reported none of the available comorbidities (n = 971), there remained an independent correlation of age with ln IL-6 and ln sTNFR-1. After adjusting for age, sex, and number of reported comorbidities, there was a statistically significant correlation between increased ln IL-6, ln IL-10, ln sTNFR-1, and ln hs-CRP with death. This study highlights the existence of a correlation between serum biomarkers of inflammation and aging, not only in the whole population, but also in the smaller subset who reported no comorbidities, confirming the existence of a presence of low-grade inflammation in aging, even in healthy elders. We also highlight the existence of a correlation between inflammatory markers and overall mortality. Future studies should address a possible threshold of systemic inflammation where mortality significantly increases, as well as explore the effectiveness of anti-inflammatory treatments on morbidity and mortality in healthy aging subjects.


Asunto(s)
Proteína C-Reactiva , Proteína Antagonista del Receptor de Interleucina 1 , Humanos , Anciano , Proteína C-Reactiva/análisis , Interleucina-10 , Interleucina-6 , Factor de Crecimiento Transformador beta1 , Jubilación , Estudios Transversales , Inflamación , Biomarcadores , Receptores del Factor de Necrosis Tumoral
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.
Exp Hematol ; 114: 1-8, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35908627

RESUMEN

The discovery of hematopoietic stem cells (HSCs) heterogeneity has had major implications for investigations of hematopoietic stem cell disorders, clonal hematopoiesis, and HSC aging. More recent studies of the heterogeneity of HSCs' organelles have begun to provide additional insights into HSCs' behavior with far-reaching ramifications for the mechanistic understanding of aging of HSCs and stem cell-derived diseases. Mitochondrial heterogeneity has been explored to expose HSC subsets with distinct properties and functions. Here we review some of the recent advances in these lines of studies that challenged the classic view of glycolysis in HSCs and led to the identification of lysosomes as dynamic pivotal switches in controlling HSC quiescence versus activation beyond their function in autophagy.


Asunto(s)
Células Madre Hematopoyéticas , Mitocondrias , Glucólisis , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo
5.
STAR Protoc ; 3(4): 101828, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595934

RESUMEN

Mitochondrial membrane potential (MMP) segregates functionally distinct subsets within highly purified hematopoietic stem cells (HSCs). Here, we detail a protocol for FACS isolation of MMP sub-fractions of phenotypically defined mouse and human HSCs. These steps are followed by high-/super-resolution immunofluorescence microscopy of HSCs' lysosomes. While the protocol describes the isolation of quiescent HSCs, which are the most potent subsets, it could also be applied to other HSC subsets. This protocol overcomes some experimental challenges associated with low HSC numbers. For complete details on the use and execution of this protocol, please refer to Liang et al. (2020) and Qiu et al. (2021).


Asunto(s)
Células Madre Hematopoyéticas , Humanos , Animales , Ratones , Citometría de Flujo/métodos
6.
Blood Adv ; 5(6): 1605-1616, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33710339

RESUMEN

Hematopoietic cell transplantation is a critical curative approach for many blood disorders. However, obtaining grafts with sufficient numbers of hematopoietic stem cells (HSCs) that maintain long-term engraftment remains challenging; this is due partly to metabolic modulations that restrict the potency of HSCs outside of their native environment. To address this, we focused on mitochondria. We found that human HSCs are heterogeneous in their mitochondrial activity as measured by mitochondrial membrane potential (MMP) even within the highly purified CD34+CD38-CD45RA-CD90+CD49f+ HSC population. We further found that the most potent HSCs exhibit the lowest mitochondrial activity in the population. We showed that the frequency of long-term culture initiating cells in MMP-low is significantly greater than in MMP-high CD34+CD38-CD45RA-CD90+ (CD90+) HSCs. Notably, these 2 populations were distinct in their long-term repopulating capacity when transplanted into immunodeficient mice. The level of chimerism 7 months posttransplantation was >50-fold higher in the blood of MMP-low relative to MMP-high CD90+ HSC recipients. Although more than 90% of both HSC subsets were in G0, MMP-low CD90+ HSCs exhibited delayed cell-cycle priming profile relative to MMP-high HSCs. These functional differences were associated with distinct mitochondrial morphology; MMP-low in contrast to MMP-high HSCs contained fragmented mitochondria. Our findings suggest that the lowest MMP level selects for the most potent, likely dormant, stem cells within the highly purified HSC population. These results identify a new approach for isolating highly potent human HSCs for further clinical applications. They also implicate mitochondria in the intrinsic regulation of human HSC quiescence and potency.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Animales , Ciclo Celular , Quimerismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Mitocondrias
7.
Cell Stem Cell ; 28(3): 374-377, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33667358

RESUMEN

The depth of quiescence in hematopoietic stem cells (HSCs) dictates their potency and is sensitive to metabolic perturbations. Recent evidence suggests that lysosomal functions distinct from autophagic processes are pivotal in regulating quiescence versus activation by potential control of the access to a nutrient reservoir required for HSC activation.


Asunto(s)
Células Madre Hematopoyéticas , Lisosomas , Autofagia , División Celular
8.
Exp Hematol ; 95: 13-22, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33440185

RESUMEN

Erythropoiesis is an intricate process starting in hematopoietic stem cells and leading to the daily production of 200 billion red blood cells (RBCs). Enucleation is a greatly complex and rate-limiting step during terminal maturation of mammalian RBC production involving expulsion of the nucleus from the orthochromatic erythroblasts, resulting in the formation of reticulocytes. The dynamic enucleation process involves many factors ranging from cytoskeletal proteins to transcription factors to microRNAs. Lack of optimum terminal erythroid maturation and enucleation has been an impediment to optimum RBC production ex vivo. Major efforts in the past two decades have exposed some of the mechanisms that govern the enucleation process. This review focuses in detail on mechanisms implicated in enucleation and discusses the future perspectives of this fascinating process.


Asunto(s)
Núcleo Celular , Eritroblastos/ultraestructura , Eritrocitos/ultraestructura , Eritropoyesis , Reticulocitos/ultraestructura , Animales , Aves/sangre , Calcio/fisiología , Cromatina/ultraestructura , Ensayo de Unidades Formadoras de Colonias , Biología Computacional , Citocinas/fisiología , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Eritroblastos/citología , Eritrocitos/citología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Mamíferos/sangre , Ratones , MicroARNs/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Hormona Tiroidea/fisiología , Proteínas Represoras/fisiología , Reticulocitos/citología , Factores de Transcripción/fisiología , Vesículas Transportadoras/fisiología , Saco Vitelino/citología , Proteínas de Unión al GTP rho/fisiología
9.
Cell Stem Cell ; 26(3): 359-376.e7, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109377

RESUMEN

Quiescence is a fundamental property that maintains hematopoietic stem cell (HSC) potency throughout life. Quiescent HSCs are thought to rely on glycolysis for their energy, but the overall metabolic properties of HSCs remain elusive. Using combined approaches, including single-cell RNA sequencing (RNA-seq), we show that mitochondrial membrane potential (MMP) distinguishes quiescent from cycling-primed HSCs. We found that primed, but not quiescent, HSCs relied readily on glycolysis. Notably, in vivo inhibition of glycolysis enhanced the competitive repopulation ability of primed HSCs. We further show that HSC quiescence is maintained by an abundance of large lysosomes. Repression of lysosomal activation in HSCs led to further enlargement of lysosomes while suppressing glucose uptake. This also induced increased lysosomal sequestration of mitochondria and enhanced the competitive repopulation ability of primed HSCs by over 90-fold in vivo. These findings show that restraining lysosomal activity preserves HSC quiescence and potency and may be therapeutically relevant.


Asunto(s)
Células Madre Hematopoyéticas , Mitocondrias , División Celular , Glucólisis , Células Madre Hematopoyéticas/metabolismo , Lisosomas , Mitocondrias/metabolismo
10.
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
11.
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
12.
Nat Commun ; 9(1): 5200, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518851

RESUMEN

BRD4 assembles transcriptional machinery at gene super-enhancer regions and governs the expression of genes that are critical for cancer progression. However, it remains unclear whether BRD4-mediated gene transcription is required for tumor cells to develop drug resistance. Our data show that prolonged treatment of luminal breast cancer cells with AKT inhibitors induces FOXO3a dephosphorylation, nuclear translocation, and disrupts its association with SirT6, eventually leading to FOXO3a acetylation as well as BRD4 recognition. Acetylated FOXO3a recognizes the BD2 domain of BRD4, recruits the BRD4/RNAPII complex to the CDK6 gene promoter, and induces its transcription. Pharmacological inhibition of either BRD4/FOXO3a association or CDK6 significantly overcomes the resistance of luminal breast cancer cells to AKT inhibitors in vitro and in vivo. Our study reports the involvement of BRD4/FOXO3a/CDK6 axis in AKTi resistance and provides potential therapeutic strategies for treating resistant breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/metabolismo , Acetilación/efectos de los fármacos , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Femenino , Proteína Forkhead Box O3/genética , Regulación de la Expresión Génica , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Ratones , Ratones Desnudos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Oxadiazoles/administración & dosificación , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Inhibidores de Proteínas Quinasas/administración & dosificación , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Pirimidinas/administración & dosificación , Pirroles/administración & dosificación , Sirtuinas/genética , Sirtuinas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética
13.
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
14.
Curr Top Dev Biol ; 127: 23-47, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29433739

RESUMEN

Stem cells self-renew and differentiate to generate all tissues and cells in the body. Stem cell health promotes adaptive responses to tissue damage or loss and is essential for tissue regeneration with age. In the past decade, the evolutionarily conserved transcription factors FOXO with known functions in promoting healthy aging have emerged as key regulators of stem cell homeostasis in various tissues, including blood, neural, and muscle stem cells. Aberrant FOXO functions have been implicated in a variety of disorders including neurodegenerative, blood, cancer, and diabetes some of which are fostered by abnormal stem cell function. As discussed in this chapter, at least in some stem cells FOXO regulatory mechanisms and applied functions follow a complex set of rules distinct from that operating in progenitor cell populations and in cultured cell lines. Elucidating the exact nature of FOXO properties in stem cells will be critical for identifying and targeting aberrant FOXO-mediated mechanisms that promote stem cell-derived disease specifically with age.


Asunto(s)
Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Homeostasis/genética , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Factores de Transcripción Forkhead/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo
16.
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
17.
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
18.
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
19.
Cell Cycle ; 15(6): 861-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26929388

RESUMEN

Transcription factors FOXOs (1, 3, 4) are essential for the maintenance of haematopoietic stem cells. FOXOs are evolutionary conserved substrates of the AKT serine threonine protein kinase that are also phosphorylated by several kinases other than AKT. Specifically, phosphorylation by AKT is known to result in the cytosolic localization of FOXO and subsequent inhibition of FOXO transcriptional activity. In addition to phosphorylation, FOXOs are regulated by a number of other post-translational modifications including acetylation, methylation, redox modulation, and ubiquitination that altogether determine these factors' output. Cumulating evidence raises the possibility that in stem cells, including in haematopoietic stem cells, AKT may not be the dominant regulator of FOXO. To address this question in more detail, we examined gene expression, subcellular localization, and response to AKT inhibition of FOXO1 and FOXO3, the main FOXO expressed in HSPCs (haematopoietic stem and progenitor cells). Here we show that while FOXO1 and FOXO3 transcripts are expressed at similar levels, endogenous FOXO3 protein is mostly nuclear compared to the cytoplasmic localization of FOXO1 in HSPCs. Furthermore, inhibition of AKT does not enhance nuclear localization of FOXO1 nor FOXO3. Nonetheless AKT inhibition in the context of loss of NAD-dependent SIRT1 deacetylase modulates FOXO3 localization in HSPCs. Together, these data suggest that FOXO3 is more active than FOXO1 in primitive haematopoietic stem and multipotent progenitor cells. In addition, they indicate that upstream regulators other than AKT, such as SIRT1, maintain nuclear FOXO localization and activity in HSPCs.


Asunto(s)
Proteína Forkhead Box O1/genética , Proteína Forkhead Box O3/genética , Células Madre Hematopoyéticas/metabolismo , ARN Mensajero/genética , Sirtuina 1/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Núcleo Celular/metabolismo , Cromonas/farmacología , Citosol/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fosforilación , Cultivo Primario de Células , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Sirtuina 1/deficiencia
20.
Matrix Biol ; 52-54: 88-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26610678

RESUMEN

We have recently demonstrated that fibrillin-1 assemblies regulate the fate of skeletal stem cells (aka, mesenchymal stem cells [MSCs]) by modulating TGFß activity within the microenvironment of adult bone marrow niches. Since MSCs can also influence hematopoietic stem cell (HSC) activities, here we investigated adult hematopoiesis in mice with Cre-mediated inactivation of the fibrillin-1 (Fbn1) gene in the mesenchyme of the forming limbs (Fbn1(Prx1-/-) mice). Analyses of 3-month-old Fbn1(Prx1-/-) mice revealed a statistically significant increase of circulating red blood cells, which a differentiation assay correlated with augmented erythropoiesis. This finding, together with evidence of fibrillin-1 deposition in erythroblastic niches, supported the notion that this extracellular matrix protein normally restricts differentiation of erythroid progenitors. Whereas flow cytometry measurements identified a decreased HSC frequency in mutant relative to wild type mice, no appreciable differences were noted with regard to the relative abundance and differentiation potential of myeloid progenitor cells. Together these findings implied that fibrillin-1 normally promotes HSC expansion but does not influence cell lineage commitment. Since local TGFß hyperactivity has been associated with abnormal osteogenesis in Fbn1(Prx1-/-) mice, 1-month-old mutant and wild type animals were systemically treated for 8weeks with either a pan-TGF-ß-neutralizing antibody or an antibody of the same IgG1 isotype. The distinct outcomes of these pharmacological interventions strongly suggest that fibrillin-1 differentially modulates TGFß activity in HSC vs. erythroid niches.


Asunto(s)
Células Madre Adultas/metabolismo , Células de la Médula Ósea/citología , Fibrilina-1/genética , Hematopoyesis , Animales , Diferenciación Celular , Fibrilina-1/metabolismo , Citometría de Flujo , Técnicas de Inactivación de Genes , Humanos , Ratones , Transducción de Señal , Nicho de Células Madre , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA