Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.176
Filtrar
Más filtros











Intervalo de año de publicación
1.
Elife ; 122024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38809590

RESUMEN

Hematopoietic stem cells emerge in the embryo from an aortic-derived tissue called the hemogenic endothelium (HE). The HE appears to give birth to cells of different nature and fate but the molecular principles underlying this complexity are largely unknown. Here we show, in the zebrafish embryo, that two cell types emerge from the aortic floor with radically different morphodynamics. With the support of live imaging, we bring evidence suggesting that the mechanics underlying the two emergence types rely, or not, on apicobasal polarity establishment. While the first type is characterized by reinforcement of apicobasal polarity and maintenance of the apical/luminal membrane until release, the second type emerges via a dynamic process reminiscent of trans-endothelial migration. Interfering with Runx1 function suggests that the balance between the two emergence types depends on tuning apicobasal polarity at the level of the HE. In support of this and unexpectedly, we show that Pard3ba - one of the four Pard3 proteins expressed in the zebrafish - is sensitive to interference with Runx1 activity, in aortic endothelial cells. This supports the idea of a signaling cross talk controlling cell polarity and its associated features, between aortic and hemogenic cells. In addition, using new transgenic fish lines that express Junctional Adhesion Molecules and functional interference, we bring evidence for the essential role of ArhGEF11/PDZ-RhoGEF in controlling the HE-endothelial cell dynamic interface, including cell-cell intercalation, which is ultimately required for emergence completion. Overall, we highlight critical cellular and dynamic events of the endothelial-to-hematopoietic transition that support emergence complexity, with a potential impact on cell fate.


In mammals and other animals with backbones, the cells that will make up blood and immune cells are generated during a very narrow timeframe in embryonic development. These cells, called hematopoietic stem cells and progenitors (or HSPCs for short), emerge from tissue known as hemogenic endothelium that makes up the floor of early blood vessels. For HPSCs to eventually specialise into different types of blood and immune cells, they require diverse migratory and homing properties that, ultimately, will determine the specific type of functions they exert. An important question for scientists studying the development of different blood and immune cell types is when this commitment to functional diversity is established. It could, for example, arise due to cells in the hemogenic endothelium having different origins. Alternatively, the signals that generate hemogenic endothelium cells could be responsible. It is also possible that both explanations are true, and that having different mechanisms involved ensures diversity in populations of HSPCs. To investigate differences between the HSPCs emerging from the hemogenic endothelium, Torcq et al. studied zebrafish embryos that had been modified so that one of the proteins involved in sensing cell polarity ­ where the top and bottom of the cell are located ­ was fluorescent. Live imaging of the embryos showed that two types of cells, with striking differences in morphology, emerge from the hemogenic tissue. In addition, one cell type displays the same polarity as the other vessel cells, whereas the other does not. Torcq et al. also present evidence suggesting that the signals responsible for controlling this cell polarity are provided by surrounding blood vessel cells, supporting the idea of an interplay between the different cell types. The finding that two different cell types emerge from the hemogenic endothelium, reveals a potential new source of diversity in HSPCs. Ultimately, this is expected to contribute to their functional complexity, resulting in both long-term stem cells that retain their full regenerative potential into adulthood and more specialized blood and immune cells.


Asunto(s)
Polaridad Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Células Madre Hematopoyéticas , Proteínas de Pez Cebra , Pez Cebra , Pez Cebra/embriología , Animales , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemangioblastos/metabolismo , Hemangioblastos/citología , Hemangioblastos/fisiología , Embrión no Mamífero/metabolismo , Animales Modificados Genéticamente
3.
Nat Aging ; 4(4): 510-526, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38632351

RESUMEN

DNA damage contributes to the aging of hematopoietic stem cells (HSCs), yet the underlying molecular mechanisms are not fully understood. In this study, we identified a heterogeneous functional role of microcephalin (MCPH1) in the nucleus and cytoplasm of mouse HSCs. In the nucleus, MCPH1 maintains genomic stability, whereas in the cytoplasm, it prevents necroptosis by binding with p-RIPK3. Aging triggers MCPH1 translocation from cytosol to nucleus, reducing its cytoplasmic retention and leading to the activation of necroptosis and deterioration of HSC function. Mechanistically, we found that KAT7-mediated lysine acetylation within the NLS motif of MCPH1 in response to DNA damage facilitates its nuclear translocation. Targeted mutation of these lysines inhibits MCPH1 translocation and, consequently, compromises necroptosis. The dysfunction of necroptosis signaling, in turn, improves the function of aged HSCs. In summary, our findings demonstrate that DNA damage-induced redistribution of MCPH1 promotes HSC aging and could have broader implications for aging and aging-related diseases.


Asunto(s)
Daño del ADN , Necroptosis , Animales , Ratones , Envejecimiento/genética , Daño del ADN/genética , Inestabilidad Genómica , Células Madre Hematopoyéticas/fisiología , Translocación Genética
4.
Front Immunol ; 15: 1339977, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38524139

RESUMEN

With the continuous development of nuclear technology, the radiation exposure caused by radiation therapy is a serious health hazard. It is of great significance to further develop effective radiation countermeasures. B cells easily succumb to irradiation exposure along with immunosuppressive response. The approach to ameliorate radiation-induced B cell damage is rarely studied, implying that the underlying mechanisms of B cell damage after exposure are eager to be revealed. Recent studies suggest that Notch signaling plays an important role in B cell-mediated immune response. Notch signaling is a critical regulator for B cells to maintain immune function. Although accumulating studies reported that Notch signaling contributes to the functionality of hematopoietic stem cells and T cells, its role in B cells is scarcely appreciated. Presently, we discussed the regulation of Notch signaling on B cells under radiation exposure to provide a scientific basis to prevent radiation-induced B cell damage.


Asunto(s)
Fenómenos Biológicos , Exposición a la Radiación , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Transducción de Señal/fisiología
5.
Biochem Biophys Res Commun ; 703: 149686, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38367513

RESUMEN

Transforming growth factor ß1 (TGFB1) refers to a pleiotropic cytokine exerting contrasting roles in hematopoietic stem cells (HSCs) functions in vitro and in vivo. However, the understanding of hematopoiesis in vivo, when TGFB1 is constantly deactivated, is still unclear, mainly due to significant embryonic lethality and the emergence of a fatal inflammatory condition, which makes doing these investigations challenging. Our study aims to find the specific role of TGFB1 in regulating hematopoiesis in vivo. We engineered mice strains (Vav1 or Mx1 promoter-driven TGFB1 knockout) with conditional knockout of TGFB1 to study its role in hematopoiesis in vivo. In fetal and adult hematopoiesis, TGFB1 KO mice displayed deficiency and decreased self-renewal capacity of HSCs with myeloid-biased differentiation. The results were different from the regulating role of TGFB1 in vitro. Additionally, our results showed that TGFB1 deficiency from fetal hematopoiesis stage caused more severe defect of HSCs than in the adult stage. Mechanistically, our findings identified TGFB1-SOX9-FOS/JUNB/TWIST1 signal axis as an essential regulating pathway in HSCs homeostasis. Our study may provide a scientific basis for clinical HSC transplantation and expansion.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Factor de Crecimiento Transformador beta1 , Animales , Ratones , Diferenciación Celular , Citocinas/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factor de Crecimiento Transformador beta1/metabolismo
7.
Toxicology ; 499: 153655, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37871686

RESUMEN

The hematopoietic factor granulocyte macrophage-colony stimulating factor (GM-CSF) has been identified via its capacity to promote bone marrow progenitors' development and differentiation into granulocytes and macrophages. Extensive pre-clinical research has established its promise as a critical therapeutic target in an assortment of inflammatory and autoimmune disorders. Despite the broad literature on GM-CSF as hematopoietic of stem cells, the cyto/geno protective aspects remain unknown. This study aimed to assess the cyto/geno protective possessions of GM-CSF on cypermethrin-induced cellular toxicity on HFF-1 cells as an in vitro model. In pre-treatment culture, cells were exposed to various GM-CSF concentrations (5, 10, 20, and 40 ng/mL) with cypermethrin at IC50 (5.13 ng/mL). Cytotoxicity, apoptotic rates, and genotoxicity were measured using the MTT, Annexin V-FITC/PI staining via flow-cytometry, and the comet assay. Cypermethrin at 5.13 ng/mL revealed cytotoxicity, apoptosis, oxidative stress, and genotoxicity while highlighting GM-CSF's protective properties on HFF-1. GM-CSF markedly attenuated cypermethrin-induced apoptotic cell death (early and late apoptotic rates). GM-CSF considerably regulated oxidative stress and genotoxicity by reducing the ROS and LPO levels, maintaining the status of GSH and activity of SOD, and suppressing genotoxicity in the comet assay parameters. Therefore, GM-CSF could be promising as an antioxidant, anti-apoptotic, genoprotective and cytomodulating agent.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Células Madre Hematopoyéticas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/fisiología , Granulocitos , Fibroblastos
8.
Nat Commun ; 14(1): 6402, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880234

RESUMEN

Lympho-hematopoiesis is regulated by cytokines; however, it remains unclear how cytokines regulate hematopoietic stem cells (HSCs) to induce production of lymphoid progenitors. Here, we show that in mice whose CXC chemokine ligand 12 (CXCL12) is deleted from half HSC niche cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, HSCs migrate from CXCL12-deficient niches to CXCL12-intact niches. In mice whose CXCL12 is deleted from all Ebf3+/leptin receptor (LepR)+ CAR cells, HSCs are markedly reduced and their ability to generate B cell progenitors is reduced compared with that to generate myeloid progenitors even when transplanted into wild-type mice. Additionally, CXCL12 enables the maintenance of B lineage repopulating ability of HSCs in vitro. These results demonstrate that CAR cell-derived CXCL12 attracts HSCs to CAR cells within bone marrow and plays a critical role in the maintenance of HSCs, especially lymphoid-biased or balanced HSCs. This study suggests an additional mechanism by which cytokines act on HSCs to produce B cells.


Asunto(s)
Quimiocinas CXC , Células Madre Hematopoyéticas , Ratones , Animales , Ligandos , Células Madre Hematopoyéticas/fisiología , Médula Ósea , Hematopoyesis , Quimiocina CXCL12 , Nicho de Células Madre , Factores de Transcripción
9.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(4): 1229-1232, 2023 Aug.
Artículo en Chino | MEDLINE | ID: mdl-37551503

RESUMEN

The homing and engraftment of hematopoietic stem cells (HSC) into bone marrow is the first critical step for successful clinical hematopoietic stem cell transplantation (HSCT). SDF-1 / CXCR4 is considered to be a very promising target to promote HSC homing. In recent years, with the in-depth research on the HSC homing, a variety of new strategies for promoting HSC homing and engraftment have been explored, such as nuclear hormone receptor, histone deacetylase inhibitor, prostaglandin and metabolic regulation, so as to increase the success rate of HSCT and improve the survival of patients. In this review, the recent research advances in the mechanism of HSC homing and strategies to promote HSC homing and engraftment were summarized and discussed.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Células Madre Hematopoyéticas/fisiología , Médula Ósea , Regulación de la Expresión Génica , Prostaglandinas/metabolismo
10.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(4): 1242-1246, 2023 Aug.
Artículo en Chino | MEDLINE | ID: mdl-37551506

RESUMEN

Bone marrow macrophage is an important component of bone marrow microenvironment, which is closely related to hematopoietic regulation and hematopoietic stem cell transplantation(HSCT). Recent studies have shown that bone marrow macrophage is an important part of hematopoietic stem cell niche, which can help regulate the mobilization and function of hematopoietic stem/progenitor cells. After HSCT, the microenvironment of bone marrow is damaged and a large number of macrophages infiltrate into the bone marrow. Regulating the macrophage-related signal pathways can promote the recovery of hematopoiesis and the reconstruction of hematopoietic function. Co-culture of macrophages and hematopoietic stem cells (HSC) in vitro significantly increased the number of HSCs and their ability of clone formation, which suggests that macrophages play an important role in the regulation of hematopoiesis in the hematopoietic microenvironment of bone marrow. This paper reviews the recent research progress on the role of macrophages in bone marrow hematopoietic microenvironment.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Humanos , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/fisiología , Hematopoyesis/fisiología , Nicho de Células Madre , Macrófagos/metabolismo
11.
Cell Stem Cell ; 30(6): 781-799.e9, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267914

RESUMEN

Somatic mutations commonly occur in hematopoietic stem cells (HSCs). Some mutant clones outgrow through clonal hematopoiesis (CH) and produce mutated immune progenies shaping host immunity. Individuals with CH are asymptomatic but have an increased risk of developing leukemia, cardiovascular and pulmonary inflammatory diseases, and severe infections. Using genetic engineering of human HSCs (hHSCs) and transplantation in immunodeficient mice, we describe how a commonly mutated gene in CH, TET2, affects human neutrophil development and function. TET2 loss in hHSCs produce a distinct neutrophil heterogeneity in bone marrow and peripheral tissues by increasing the repopulating capacity of neutrophil progenitors and giving rise to low-granule neutrophils. Human neutrophils that inherited TET2 mutations mount exacerbated inflammatory responses and have more condensed chromatin, which correlates with compact neutrophil extracellular trap (NET) production. We expose here physiological abnormalities that may inform future strategies to detect TET2-CH and prevent NET-mediated pathologies associated with CH.


Asunto(s)
Dioxigenasas , Neutrófilos , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas , Células Madre Hematopoyéticas/fisiología , Médula Ósea , Hematopoyesis/genética , Mutación , Proteínas de Unión al ADN/genética , Dioxigenasas/genética
12.
Int J Mol Sci ; 24(9)2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37175857

RESUMEN

Megakaryocytes are the main members of the hematopoietic system responsible for regulating vascular homeostasis through their progeny platelets, which are generally known for maintaining hemostasis. Megakaryocytes are characterized as large polyploid cells that reside in the bone marrow but may also circulate in the vasculature. They are generated directly or through a multi-lineage commitment step from the most primitive progenitor or Hematopoietic Stem Cells (HSCs) in a process called "megakaryopoiesis". Immature megakaryocytes enter a complicated development process defined as "thrombopoiesis" that ultimately results in the release of extended protrusions called proplatelets into bone marrow sinusoidal or lung microvessels. One of the main mediators that play an important modulatory role in hematopoiesis and hemostasis is nitric oxide (NO), a free radical gas produced by three isoforms of nitric oxide synthase within the mammalian cells. In this review, we summarize the effect of NO and its signaling on megakaryopoiesis and thrombopoiesis under both physiological and pathophysiological conditions.


Asunto(s)
Megacariocitos , Óxido Nítrico , Animales , Megacariocitos/fisiología , Plaquetas , Trombopoyesis , Células Madre Hematopoyéticas/fisiología , Mamíferos
13.
Nat Commun ; 14(1): 2018, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037837

RESUMEN

Aging associated defects within stem cell-supportive niches contribute towards age-related decline in stem cell activity. However, mechanisms underlying age-related niche defects, and whether restoring niche function can improve stem cell fitness, remain unclear. Here, we sought to determine whether aged blood stem cell function can be restored by rejuvenating their supportive niches within the bone marrow (BM). We identify Netrin-1 as a critical regulator of BM niche cell aging. Niche-specific deletion of Netrin-1 induces premature aging phenotypes within the BM microenvironment, while supplementation of aged mice with Netrin-1 rejuvenates aged niche cells and restores competitive fitness of aged blood stem cells to youthful levels. We show that Netrin-1 plays an essential role in maintaining active DNA damage responses (DDR), and that aging-associated decline in niche-derived Netrin-1 results in DNA damage accumulation within the BM microenvironment. We show that Netrin-1 supplementation is sufficient to resolve DNA damage and restore regenerative potential of the aged BM niche and blood stem cells to endure serial chemotherapy regimens.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Ratones , Netrina-1/genética , Células Madre Hematopoyéticas/fisiología , Células de la Médula Ósea , Envejecimiento/genética , Nicho de Células Madre
14.
Stem Cell Res Ther ; 14(1): 73, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37038215

RESUMEN

Hematopoietic stem cells (HSCs) are the common and essential precursors of all blood cells, including immune cells, and they are responsible for the lifelong maintenance and damage repair of blood tissue homeostasis. The vast majority (> 95%) of HSCs are in a resting state under physiological conditions and are only activated to play a functional role under stress conditions. This resting state affects their long-term survival and is also closely related to the lifelong maintenance of hematopoietic function; however, abnormal changes may also be an important factor leading to the decline of immune function in the body and the occurrence of diseases in various systems. While the importance of resting HSCs has attracted increasing research attention, our current understanding of this topic remains insufficient, and the direction of clinical targeted treatments is unclear. Here, we describe the functions of HSCs, analyze the regulatory mechanisms that affect their resting state, and discuss the relationship between resting HSCs and different diseases, with a view to providing guidance for the future clinical implementation of related targeted treatments.


Asunto(s)
Células Sanguíneas , Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Diferenciación Celular/fisiología
15.
J Clin Lab Anal ; 37(6): e24872, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36972475

RESUMEN

AIM: Hematopoietic stem cells are the origin of all hematopoietic cells. They have the self-renewal ability and can differentiate into various blood cells. In physiological state, most of the hematopoietic stem cells are dormant, and only a few cells proliferate to maintain hematopoietic homeostasis. METHODS: This precise steady-state maintenance is regulated by complex mechanisms. Bone marrow adipocytes make up half of all cells in the bone marrow cavity, a feature that has attracted the attention of researchers from multiple fields. The adipocyte density within marrow increases during aging and obesity. RESULTS: Recent studies have shown that bone marrow adipocytes play important roles in regulating hematopoiesis, but the effects of bone marrow adipocytes on hematopoiesis are often conflicting. Bone marrow adipocytes, participating in the formation of bone marrow hematopoietic microenvironment, influence hematopoiesis positively or negatively. In addition, other adipose tissue, especially white adipose tissue, also regulates hematopoiesis. CONCLUSION: In this review, we describe the role of adipose tissue in hematological malignancies, which may be useful for understanding hematopoiesis and the pathogenesis of related diseases.


Asunto(s)
Tejido Adiposo , Células de la Médula Ósea , Humanos , Células de la Médula Ósea/fisiología , Médula Ósea , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología
16.
Int J Radiat Biol ; 99(10): 1483-1494, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36912588

RESUMEN

PURPOSE: The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION: In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, ß-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Médula Ósea/efectos de la radiación , Glándulas Salivales/efectos de la radiación , Citocinas/metabolismo
17.
Small Methods ; 7(6): e2201300, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36843214

RESUMEN

The sympathetic nervous system (SNS) of the bone marrow regulates the regeneration and mobilization of hematopoietic stem cells. Chemotherapy can damage bone marrow SNS, which impairs hematopoietic regeneration and aggravates hematologic toxicities. This leads to long-term bone marrow niche damage and increases mortality in patients undergoing chemotherapy. Electrical neuromodulation has been used to improve functional recovery after peripheral nerve injury. This study demonstrates that electrical sympathetic neuromodulation (ESN) of bone marrow can protect the bone marrow niche from chemotherapy-induced injury. Using carboplatin-treated rats, the SNS via the sciatic nerve innervating the femoral marrow with the effective protocol for bone marrow sympathetic activation is electrically stimulated. ESN can mediate several hematopoietic stem cells maintenance factors and promote hematopoietic regeneration after chemotherapy. It also activates adrenergic signals and reduces the release of pro-inflammatory cytokines, particularly interleukin-1 ß, which contribute to chemotherapy-related nerve injury. Consequently, the severity of chemotherapy-related leukopenia, thrombocytopenia, and mortality can be reduced by ESN. As a result, in contrast to current drug-based treatment, such as granulocyte colony-stimulating factor, ESN can be a disruptive adjuvant treatment by protecting and modulating bone marrow function to reduce hematologic toxicity during chemotherapy.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Ratas , Animales , Células Madre Hematopoyéticas/fisiología , Citocinas/farmacología , Células de la Médula Ósea , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos/uso terapéutico
18.
Blood ; 141(20): 2483-2492, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-36787502

RESUMEN

Hematopoietic stem cells (HSCs) are the ultimate source of blood and immune cells, and transplantation reveals their unique potential to regenerate all blood lineages lifelong. HSCs are considered a quiescent reserve population under homeostatic conditions, which can be rapidly activated by perturbations to fuel blood regeneration. In accordance with this concept, inflammation and loss of blood cells were reported to stimulate the proliferation of HSCs, which is associated with a decline in their transplantation potential. To investigate the contribution of primitive HSCs to the hematopoietic stress response in the native environment, we use fate mapping and proliferation tracking mouse models. Although primitive HSCs were robustly activated by severe myeloablation, they did not contribute to the regeneration of mature blood cells in response to prototypic hematopoietic emergencies, such as acute inflammation or blood loss. Even chronic inflammatory stimulation, which triggered vigorous HSC proliferation, only resulted in a weak contribution of HSCs to mature blood cell production. Thus, our data demonstrate that primitive HSCs do not participate in the hematopoietic recovery from common perturbations and call for the reevaluation of the concept of HSC-driven stress responses.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Animales , Ratones , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Regeneración/fisiología , Inflamación
19.
Blood ; 142(6): 533-542, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-36800569

RESUMEN

With aging, hematopoietic stem cells (HSCs) have an impaired ability to regenerate, differentiate, and produce an entire repertoire of mature blood and immune cells. Owing to dysfunctional hematopoiesis, the incidence of hematologic malignancies increases among elderly individuals. Here, we provide an update on HSC-intrinsic and -extrinsic factors and processes that were recently discovered to contribute to the functional decline of HSCs during aging. In addition, we discuss the targets and timing of intervention approaches to maintain HSC function during aging and the extent to which these same targets may prevent or delay transformation to hematologic malignancies.


Asunto(s)
Neoplasias Hematológicas , Leucemia , Humanos , Anciano , Senescencia Celular , Envejecimiento , Células Madre Hematopoyéticas/fisiología , Leucemia/terapia , Hematopoyesis/fisiología , Neoplasias Hematológicas/terapia
20.
Int J Radiat Oncol Biol Phys ; 116(5): 1163-1174, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36792018

RESUMEN

PURPOSE: Victims of acute radiation exposure are susceptible to hematopoietic toxicity due to bone marrow damage and loss of mature blood elements. Here, we evaluated cord blood-derived endothelial progenitor cells (CB-EPCs) as a potential cellular therapy for mitigation of hematologic acute radiation syndrome. CB-EPCs express endothelial cell markers and maintain their growth characteristics beyond 10+ passages without diminishing their doubling capacity. Further, CB-EPCs can be cryopreserved in vapor-phase liquid nitrogen and easily recovered for propagation, making them an attractive nonimmunogenic cellular therapy for off-the-shelf use. Importantly, we show CB-EPCs have the capacity to potently expand adult human bone marrow hematopoietic progenitor cells both in vitro and in vivo. METHODS AND MATERIALS: To demonstrate the role of CB-EPCs in promoting in vivo human immune reconstitution after irradiation, we employed a novel humanized mouse model established by transplant of CD34+ bone marrow cells from 9 unique adult organ donors into immunocompromised NSG-SGM3 mice. The response of the humanized immune system to ionizing irradiation was then tested by exposure to 1 Gy followed by subcutaneous treatment of CB-EPCs, Food and Drug Administration-approved growth factor pegfilgrastim (0.3 mg/kg), or saline. RESULTS: At day 7, total human bone marrow was decreased by 80% in irradiated controls. However, treatment with either growth factor pegfilgrastim or CB-EPCs increased recovery of total human bone marrow by 2.5-fold compared with saline. Notably, CB-EPCs also increased recovery of both human CD34+ progenitors by 5-fold and colony-forming capacity by 3-fold versus saline. Additionally, CB-EPCs promoted recovery of endogenous bone marrow endothelial cells as observed by both increased vessel area and length compared with saline. CONCLUSIONS: These findings indicate the feasibility of using humanized mice engrafted with adult bone marrow for radiation research and the development of CB-EPCs as an off-the-shelf cellular therapy for mitigation of hematologic acute radiation syndrome.


Asunto(s)
Síndrome de Radiación Aguda , Células Progenitoras Endoteliales , Trasplante de Células Madre Hematopoyéticas , Adulto , Humanos , Ratones , Animales , Médula Ósea , Células Madre Hematopoyéticas/fisiología , Sangre Fetal/metabolismo , Síndrome de Radiación Aguda/metabolismo , Células de la Médula Ósea , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA