Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.570
Filtrar
1.
PeerJ ; 12: e18054, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39314840

RESUMEN

Background: Ineffective erythropoiesis (IE) is the primary cause of anemia and associated pathologies in ß-thalassemia. The characterization of IE is imbalance of erythroid proliferation and differentiation, resulting in increased erythroblast proliferation that fails to differentiate and gives rise to enucleate RBCs. MicroRNAs (miRs) are known to play important roles in hematopoiesis. miR-155 is a multifunctional molecule involved in both normal and pathological hematopoiesis, and its upregulation is observed in patients with ß-thalassemia/HbE. However, the expression and function of miR-155, especially in ß-thalassemia, have not yet been explored. Methods: To study miR-155 expression in thalassemia, erythroblast subpopulations, CD45-CD71+Ter-119+ and CD45-CD71-Ter-119+ were collected from ß IVSII-654 thalassemic bone marrow. Additionally, a two-phase culture of mouse bone marrow erythroid progenitor cells was performed. Expression of miR-155 and predicted mRNA target genes, c-myc, bach-1 and pu-1, were determined by quantitative reverse transcription (qRT)-polymerase chain reaction (PCR) and normalized to small nucleolar RNA (snoRNA) 202 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), respectively. To investigate the effect of miR-155 expression, erythroblasts were transfected with miR-inhibitor and -mimic in order to elevate and eliminate miR-155 expression, respectively. Erythroid cell differentiation was evaluated by Wright-Giemsa staining and flow cytometry. Results: miR-155 was upregulated, both in vivo and in vitro, during erythropoiesis in ß-thalassemic mice. Our study revealed that gain- and loss of function of miR-155 were involved in erythroid proliferation and differentiation, and augmented proliferation and differentiation of thalassemic mouse erythroblasts may be associated with miR-155 upregulation. miR-155 upregulation in ß-thalassemic mice significantly increased the percentage of basophilic and polychromatic erythroblasts. Conversely, a significant decrease in percentage of basophilic and polychromatic erythroblasts was observed in ß-thalassemic mice transfected with anti-miR-155 inhibitor. We also examined the mRNA targets (c-myc, bach-1 and pu-1) of miR-155, which indicated that c-myc is a valid target gene of miR-155 that regulates erythroid differentiation. Conclusion: miR-155 regulates IE in ß-thalassemia via c-myc expression controlling erythroblast proliferation and differentiation.


Asunto(s)
Eritropoyesis , MicroARNs , Talasemia beta , MicroARNs/genética , MicroARNs/metabolismo , Eritropoyesis/genética , Animales , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/patología , Ratones , Humanos , Masculino , Diferenciación Celular , Femenino , Eritroblastos/metabolismo , Eritroblastos/patología , Transactivadores/genética , Transactivadores/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Adulto , Adolescente , Proliferación Celular , Proteínas Proto-Oncogénicas , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico
3.
Int J Cancer ; 155(11): 1928-1938, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39039820

RESUMEN

Immunotherapy, especially immune checkpoint blockade therapy, represents a major milestone in the history of cancer therapy. However, the current response rate to immunotherapy among cancer patients must be improved; thus, new strategies for sensitizing patients to immunotherapy are urgently needed. Erythroid progenitor cells (EPCs), a population of immature erythroid cells, exert potent immunosuppressive functions. As a newly recognized immunosuppressive population, EPCs have not yet been effectively targeted. In this review, we summarize the immunoregulatory mechanisms of EPCs, especially for CD45+ EPCs. Moreover, in view of the regulatory effects of EPCs on the tumor microenvironment, we propose the concept of EPC-immunity, present existing strategies for targeting EPCs, and discuss the challenges encountered in both basic research and clinical applications. In particular, the impact of existing cancer treatments on EPCs is discussed, laying the foundation for combination therapies. The aim of this review is to provide new avenues for improving the efficacy of cancer immunotherapy by targeting EPCs.


Asunto(s)
Células Precursoras Eritroides , Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Células Precursoras Eritroides/inmunología , Animales , Antígenos Comunes de Leucocito/metabolismo
4.
Cytotherapy ; 26(11): 1362-1373, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39001769

RESUMEN

BACKGROUND AIMS: Ex vivo production of red blood cells (RBCs) represents a promising alternative for transfusion medicine. Several strategies have been described to generate erythroid cell lines from different sources, including embryonic, induced pluripotent, and hematopoietic stem cells. All these approaches have in common that they require elaborate differentiation cultures whereas the yield of enucleated RBCs is inefficient. METHODS: We generated a human immortalized adult erythroid progenitor cell line derived from bone marrow CD71-positive erythroid progenitor cells (immortalized bone marrow erythroid progenitor adult, or imBMEP-A) by an inducible expression system, to shorten differentiation culture necessary for terminal erythroid differentiation. It is the first erythroid cell line that is generated from direct reticulocyte progenitors and demonstrates robust hemoglobin production in the immortalized state. RESULTS: Morphologic analysis of the immortalized cells showed that the preferred cell type of the imBMEP-A line corresponds to hemoglobin-producing basophilic erythroblasts. In addition, we were able to generate a stable cell line from a single cell clone with the triple knockout of RhAG, RhDCE and KELL. After removal of doxycycline, part of the cells differentiated into normoblasts and reticulocytes within 5-7 days. CONCLUSIONS: Our results demonstrate that the imBMEP-A cell line can serve as a stable and straightforward modifiable platform for RBC engineering in the future.


Asunto(s)
Antígenos CD , Diferenciación Celular , Células Precursoras Eritroides , Receptores de Transferrina , Humanos , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Receptores de Transferrina/metabolismo , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Eritropoyesis , Línea Celular , Eritrocitos/citología , Eritrocitos/metabolismo , Reticulocitos/citología , Reticulocitos/metabolismo
5.
Exp Hematol ; 136: 104283, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39048071

RESUMEN

Red blood cells (RBCs) comprise a critical component of the cardiovascular network, which constitutes the first functional organ system of the developing mammalian embryo. Examination of circulating blood cells in mammalian embryos revealed two distinct types of erythroid cells: large, nucleated "primitive" erythroblasts followed by smaller, enucleated "definitive" erythrocytes. This review describes the current understanding of primitive and definitive erythropoiesis gleaned from studies of mouse and human embryos and induced pluripotent stem cells (iPSCs). Primitive erythropoiesis in the mouse embryo comprises a transient wave of committed primitive erythroid progenitors (primitive erythroid colony-forming cells, EryP-CFC) in the early yolk sac that generates a robust cohort of precursors that mature in the bloodstream and enucleate. In contrast, definitive erythropoiesis has two distinct developmental origins. The first comprises a transient wave of definitive erythroid progenitors (burst-forming units erythroid, BFU-E) that emerge in the yolk sac and seed the fetal liver where they terminally mature to provide the first definitive RBCs. The second comprises hematopoietic stem cell (HSC)-derived BFU-E that terminally mature at sites colonized by HSCs particularly the fetal liver and subsequently the bone marrow. Primitive and definitive erythropoiesis are derived from endothelial identity precursors with distinct developmental origins. Although they share prototypical transcriptional regulation, primitive and definitive erythropoiesis are also characterized by distinct lineage-specific factors. The exquisitely timed, sequential production of primitive and definitive erythroid cells is necessary for the survival and growth of the mammalian embryo.


Asunto(s)
Embrión de Mamíferos , Eritropoyesis , Eritropoyesis/fisiología , Animales , Humanos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Ratones , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Saco Vitelino/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo
6.
Cells ; 13(13)2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38995000

RESUMEN

Erythropoiesis occurs first in the yolk sac as a transit "primitive" form, then is gradually replaced by the "definitive" form in the fetal liver (FL) during fetal development and in the bone marrow (BM) postnatally. While it is well known that differences exist between primitive and definitive erythropoiesis, the similarities and differences between FL and BM definitive erythropoiesis have not been studied. Here we performed comprehensive comparisons of erythroid progenitors and precursors at all maturational stages sorted from E16.5 FL and adult BM. We found that FL cells at all maturational stages were larger than their BM counterparts. We further found that FL BFU-E cells divided at a faster rate and underwent more cell divisions than BM BFU-E. Transcriptome comparison revealed that genes with increased expression in FL BFU-Es were enriched in cell division. Interestingly, the expression levels of glucocorticoid receptor Nr3c1, Myc and Myc downstream target Ccna2 were significantly higher in FL BFU-Es, indicating the role of the Nr3c1-Myc-Ccna2 axis in the enhanced proliferation/cell division of FL BFU-E cells. At the CFU-E stage, the expression of genes associated with hemoglobin biosynthesis were much higher in FL CFU-Es, indicating more hemoglobin production. During terminal erythropoiesis, overall temporal patterns in gene expression were conserved between the FL and BM. While biological processes related to translation, the tricarboxylic acid cycle and hypoxia response were upregulated in FL erythroblasts, those related to antiviral signal pathway were upregulated in BM erythroblasts. Our findings uncovered previously unrecognized differences between FL and BM definitive erythropoiesis and provide novel insights into erythropoiesis.


Asunto(s)
Médula Ósea , Eritropoyesis , Feto , Hígado , Transcriptoma , Animales , Eritropoyesis/genética , Ratones , Hígado/metabolismo , Hígado/embriología , Hígado/citología , Transcriptoma/genética , Feto/metabolismo , Feto/citología , Médula Ósea/metabolismo , Ratones Endogámicos C57BL , Regulación del Desarrollo de la Expresión Génica , Femenino , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología
7.
Eur J Haematol ; 113(4): 416-425, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38853593

RESUMEN

OBJECTIVES: Galectin-9 (Gal-9) is an immune checkpoint ligand for T-cell immunoglobulin and mucin domain 3. Although the roles of Gal-9 in regulating immune responses have been well investigated, their biological roles have yet to be fully documented. This study aimed to analyse the expression of Gal-9 bone marrow (BM) cells in C57BL/6J (B6) mice. Furthermore, the co-expression of Gal-9 with the mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) was investigated. METHODS: The BM cells in adult C57BL/6J (B6) mice were collected and analysed in vitro. RESULTS: In a flow cytometric analysis of BM cells, Gal-9 was highly expressed in c-KithiSca-1-CD34-CD71+ erythroid progenitors (EPs), whereas it was downregulated in more differentiated c-KitloCD71+TER119+ cells. Subsequently, a negative selection of CD3-B220-Sca-1-CD34-CD41-CD16/32- EPs was performed. This resulted in substantial enrichment of KithiCD71+Gal-9+ cells and erythroid colony-forming units (CFU-Es), suggesting that the colony-forming subset of EPs are included in the KithiCD71+Gal-9+ population. Furthermore, we found that EPs had lower mTOR and AMPK expression levels in Gal-9 knockout B6 mice than in wild-type B6 mice. CONCLUSIONS: These results may stimulate further investigation of the role of Gal-9 in haematopoiesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Precursoras Eritroides , Galectinas , Serina-Treonina Quinasas TOR , Animales , Galectinas/metabolismo , Galectinas/genética , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología , Ratones Endogámicos C57BL , Expresión Génica , Inmunofenotipificación , Biomarcadores , Células de la Médula Ósea/metabolismo
8.
Front Immunol ; 15: 1381919, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799424

RESUMEN

Introduction: CD8+T cell tolerance plays an important role in tumor escape. Recent studies have shown that CD45+ erythroid progenitor cells (CD45+EPCs) generated through splenic extramedullary erythropoiesis suppress tumor immunity. However, the mechanism underlying how CD45+EPCs mediate CD8+T cell tolerance remains incompletely understood and requires further research. Methods: In this study, the antigen-processing abilities of CD45+EPCs was verified through both in vitro and in vivo experiments. We have used the method of co-culture in vitro and adoptive transfer experiments in vivo to explore the effects of CD45+EPCs on CD8+T cell tolerance. RNA-sequencing analysis and blocking experiments were used to evaluate the role of ROS in the CD45+EPC mediated tolerance of CD8+T cells. Finally, we incorporated uric acid into the adoptive transfer experiments to rescue the CD45+EPC mediated tumor-promoting effect. Results and discussion: We found that CD45+EPCs take up soluble proteins, present antigenic epitopes on their surface, and induce antigen-specific CD8+T cell anergy. In addition, we found that CD45+EPC directly nitrates tyrosine within the TCR/CD8 complex via the production of reactive oxygen species and peroxynitrite, preventing CD8+ T cells from responding to their specific peptide antigens. Furthermore, uric acid treatment effectively abolished the immunosuppressive effects of CD45+EPCs during CD8+T cell adoptive transfer, thereby enhancing the anti-tumor efficacy. These results demonstrated that CD8+T cell tolerance in tumor-bearing mice is induced by CD45+EPCs. The results of this study have direct implications for tumor immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Células Precursoras Eritroides , Tolerancia Inmunológica , Animales , Linfocitos T CD8-positivos/inmunología , Ratones , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones Endogámicos C57BL , Traslado Adoptivo , Especies Reactivas de Oxígeno/metabolismo , Escape del Tumor/inmunología , Línea Celular Tumoral , Ácido Úrico
9.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605277

RESUMEN

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Asunto(s)
Proteínas de Ciclo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Eritropoyesis , Ratones Noqueados , Microcefalia , Proteína p53 Supresora de Tumor , Animales , Ratones , Anemia Macrocítica/genética , Anemia Macrocítica/patología , Anemia Macrocítica/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Microcefalia/genética , Microcefalia/patología , Mutación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
Exp Hematol ; 135: 104191, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38493949

RESUMEN

Erythropoiesis in the adult bone marrow relies on mitochondrial membrane transporters to facilitate heme and hemoglobin production. Erythrocytes in the bone marrow are produced although the differentiation of erythroid progenitor cells that originate from hematopoietic stem cells (HSCs). Whether and how mitochondria transporters potentiate HSCs and affect their differentiation toward erythroid lineage remains unclear. Here, we show that the ATP-binding cassette (ABC) transporter 10 (Abcb10), located on the inner mitochondrial membrane, is essential for HSC maintenance and erythroid-lineage differentiation. Induced deletion of Abcb10 in adult mice significantly increased erythroid progenitor cell and decreased HSC number within the bone marrow (BM). Functionally, Abcb10-deficient HSCs exhibited significant decreases in stem cell potential but with a skew toward erythroid-lineage differentiation. Mechanistically, deletion of Abcb10 rendered HSCs with excess mitochondrial iron accumulation and oxidative stress yet without alteration in mitochondrial bioenergetic function. However, impaired hematopoiesis could not be rescued through the in vivo administration of a mitochondrial iron chelator or antioxidant to Abcb10-deficient mice. Abcb10-mediated mitochondrial iron transfer is thus pivotal for the regulation of physiologic HSC potential and erythroid-lineage differentiation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Diferenciación Celular , Eritropoyesis , Células Madre Hematopoyéticas , Ratones Noqueados , Mitocondrias , Animales , Ratones , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Mitocondrias/metabolismo , Eritropoyesis/genética , Hierro/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Estrés Oxidativo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología , Ratones Endogámicos C57BL
11.
Blood Cells Mol Dis ; 106: 102829, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38278056

RESUMEN

BACKGROUND: Erythropoiesis is a complex developmental process in which a hematopoietic stem cell undergoes serial divisions and differentiates through well-defined stages to give rise to red blood cells. Over the last decades, several protocols have been developed to perform ex vivo erythroid differentiation, allowing investigation into erythropoiesis and red cell production in health and disease. RESULTS: In the current study, we compared the two commonly used protocols by assessing the differentiation kinetics, synchronisation, and cellular yield, using molecular and cellular approaches. Peripheral blood CD34+ cells were cultured in a two-phase (2P) or a four-phase (4P) liquid culture (LC) and monitored for 20 days. Both protocols could recapitulate all stages of erythropoiesis and generate reticulocytes, although to different extents. Higher proliferation and viability rates were achieved in the 4P-LC, with a higher degree of terminal differentiation and enucleation, associated with higher levels of the erythroid-specific transcription factors GATA-1, KLF-1, and TAL-1. Although the 2P-LC protocol was less efficient regarding terminal erythroid differentiation and maturation, it showed a higher yield of erythroid progenitors in the erythropoietin (EPO)-free expansion phase. CONCLUSIONS: We provide data supporting the use of one protocol or the other to study the biological processes occurring in the early or late stages of erythroid differentiation, depending on the physiological process or pathological defect under investigation in a given study.


Asunto(s)
Eritropoyetina , Células Madre Hematopoyéticas , Humanos , Diferenciación Celular , Eritrocitos , Eritropoyesis/fisiología , Antígenos CD34 , Células Precursoras Eritroides
12.
Exp Hematol ; 129: 104128, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37939833

RESUMEN

During the recent coronavirus disease 2019 (COVID-19) pandemic several patients with ß-thalassemia have been infected by severe acute respiratory syndrome coronavirus (SARS-CoV-2), and most patients were vaccinated against SARS-CoV-2. Recent studies demonstrate an impact of SARS-CoV-2 infection on the hematopoietic system. The main objective of this study was to verify the effects of exposure of erythroid precursor cells (ErPCs) from patients with ß-thalassemia to SARS-CoV-2 spike protein (S-protein) and the BNT162b2 vaccine. Erythropoietin (EPO)-cultured ErPCs have been either untreated or treated with S-protein or BNT162b2 vaccine. The employed ErPCs were from a ß-thalassemia cellular Biobank developed before the COVID-19 pandemic. The genotypes were ß+-IVSI-110/ß+-IVSI-110 (one patient),  ß039/ß+-IVSI-110 (3 patients), and ß039/ ß039 (2 patients). After treatment with S-protein or BNT162b2 for 5 days, lysates were analyzed by high performance liquid chromatography (HPLC), for hemoglobin production, and isolated RNA was assayed by RT-qPCR, for detection of globin gene expression. The main conclusions of the results obtained are that SARS-CoV-2 S-protein and BNT162b2 vaccine (a) inhibit fetal hemoglobin (HbF) production by ß-thalassemic ErPCs and (b) inhibit γ-globin mRNA accumulation. In addition, we have performed in silico studies suggesting a high affinity of S-protein to HbF. Remarkably, the binding interaction energy of fetal hemoglobin to S-protein was comparable with that of angiotensin-converting enzyme 2 (ACE2). Our results are consistent with the hypothesis of a relevant impact of SARS-CoV-2 infection and COVID-19 vaccination on the hematopoietic system.


Asunto(s)
COVID-19 , Eritropoyetina , Vacunas , Talasemia beta , Humanos , Glicoproteína de la Espiga del Coronavirus/genética , Vacuna BNT162 , Talasemia beta/genética , Células Precursoras Eritroides , Vacunas contra la COVID-19 , Hemoglobina Fetal , Pandemias , SARS-CoV-2 , Expresión Génica , Anticuerpos Antivirales
13.
Am J Hematol ; 99(1): 99-112, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37929634

RESUMEN

Human erythropoiesis is a complex process leading to the production of 2.5 million red blood cells per second. Following commitment of hematopoietic stem cells to the erythroid lineage, this process can be divided into three distinct stages: erythroid progenitor differentiation, terminal erythropoiesis, and reticulocyte maturation. We recently resolved the heterogeneity of erythroid progenitors into four different subpopulations termed EP1-EP4. Here, we characterized the growth factor(s) responsiveness of these four progenitor populations in terms of proliferation and differentiation. Using mass spectrometry-based proteomics on sorted erythroid progenitors, we quantified the absolute expression of ~5500 proteins from EP1 to EP4. Further functional analyses highlighted dynamic changes in cell cycle in these populations with an acceleration of the cell cycle during erythroid progenitor differentiation. The finding that E2F4 expression was increased from EP1 to EP4 is consistent with the noted changes in cell cycle. Finally, our proteomic data suggest that the protein machinery necessary for both oxidative phosphorylation and glycolysis is present in these progenitor cells. Together, our data provide comprehensive insights into growth factor-dependence of erythroid progenitor proliferation and the proteome of four distinct populations of human erythroid progenitors which will be a useful framework for the study of erythroid disorders.


Asunto(s)
Células Madre Hematopoyéticas , Proteómica , Humanos , Diferenciación Celular , Ciclo Celular , Eritropoyesis , Redes y Vías Metabólicas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Precursoras Eritroides
14.
Biochem Pharmacol ; 220: 116008, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38154543

RESUMEN

Recombinant human erythropoietin (rHuEPO) is a prevalent treatment for anemia in patients with chronic kidney disease. However, up to 10% of these patients exhibit EPO resistance or hyporesponsiveness, which may be caused by the depletion of erythroid progenitor cells. Thrombopoietin (TPO) has the potential to promote the growth of early progenitor cells and correct the depletion. In this study, we investigate the efficacy and the underlying mechanism of the combination therapy of TPO and EPO to EPO resistance. First, the in vivo studies suggested that intensive EPO treatment induced progenitor cell depletion in the bone marrow, where the depletion was corrected by TPO. Then, colony assays showed that EPO and TPO synergistically enhanced the burst-forming unit-erythroid (BFU-E) production but antagonistically boosted the colony-forming units of megakaryocytes (CFU-MK) production. Also, we found TPO promoted hematopoietic stem and progenitor cells (HSPCs) production, while EPO drove HSPCs toward the erythroid lineage. Additionally, EPO induced more megakaryocytic-erythroid progenitors (MEPs) toward the erythroid output. Model-based simulations indicate the efficacy of this combination therapy for treating EPO-resistant anemia in rats. In conclusion, our study demonstrated the efficacy of combination therapy in addressing EPO-resistant anemia by correcting EPO-induced erythroid progenitor depletion.


Asunto(s)
Anemia , Eritropoyetina , Animales , Humanos , Ratas , Células Precursoras Eritroides , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Células Madre Hematopoyéticas , Megacariocitos , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Trombopoyetina/farmacología , Trombopoyetina/uso terapéutico
15.
Nat Immunol ; 24(12): 2042-2052, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37919525

RESUMEN

Tumor-derived factors are thought to regulate thrombocytosis and erythrocytopenia in individuals with cancer; however, such factors have not yet been identified. Here we show that tumor cell-released kynurenine (Kyn) biases megakaryocytic-erythroid progenitor cell (MEP) differentiation into megakaryocytes in individuals with cancer by activating the aryl hydrocarbon receptor-Runt-related transcription factor 1 (AhR-RUNX1) axis. During tumor growth, large amounts of Kyn from tumor cells are released into the periphery, where they are taken up by MEPs via the transporter SLC7A8. In the cytosol, Kyn binds to and activates AhR, leading to its translocation into the nucleus where AhR transactivates RUNX1, thus regulating MEP differentiation into megakaryocytes. In addition, activated AhR upregulates SLC7A8 in MEPs to induce positive feedback. Importantly, Kyn-AhR-RUNX1-regulated MEP differentiation was demonstrated in both humanized mice and individuals with cancer, providing potential strategies for the prevention of thrombocytosis and erythrocytopenia.


Asunto(s)
Neoplasias , Trombocitosis , Animales , Ratones , Quinurenina/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Megacariocitos/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Precursoras Eritroides/metabolismo , Diferenciación Celular/fisiología , Neoplasias/metabolismo , Trombocitosis/metabolismo , Sesgo
16.
Gastric Cancer ; 26(6): 918-933, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37676622

RESUMEN

BACKGROUND AND AIMS: Specific mechanisms of lymph node (LN) metastasis in early-stage gastric cancer (GC) have not been elucidated. The role of anemia, a vital clinical feature of GC, in LN metastasis is also unclear. Since the number of erythroid progenitor cells (EPCs) is increased in chronic anemia, we investigated its association with LN metastasis in GC. METHODS: Flow cytometry and immunofluorescence analyses were performed to sort and study EPCs from the circulation and tumors of patients with stage I-III GC. The effect of these EPCs on the activation of T and B cells and on the functions of lymphatic endothelial cells (LECs) was determined, and their ability to promote LN metastasis was evaluated using a footpad-popliteal LN metastasis model based on two human adenocarcinoma GC cell lines in nude mice. The prognostic value of EPCs was also analyzed. RESULTS: The proportion of CD45- EPCs was higher in the mononuclear cells in the circulation, tumors, and LNs of GC patients with LN metastasis (N+) than in those of GC patients without LN metastasis (N0). In N+ patients, CD45- EPCs were more abundant in metastatic LNs than in non-metastatic LNs. Lymphatic vessel endothelial hyaluronan receptor 1 immunoreactivity in tumors revealed that CD45- EPCs were positively associated with nodal stages and lymph vessel density. Furthermore, CD45- EPCs increased LEC proliferation and migration through their S100A8/A9 heterodimer-induced hybrid epithelial/mesenchymal (E/M) state; however, they did not influence the invasion and tubulogenesis of LECs or T and B cell proliferation. CD45- EPCs promoted LN metastasis in vivo; the S100A8/A9 heterodimer mimicked this phenomenon. Finally, CD45- EPCs predicted the overall and disease-free survival of stage I-III GC patients after radical resection. CONCLUSIONS: The CD45- EPCs accumulated in GC tissues and metastatic LNs and promoted LN metastasis via the S100A8/9-induced hybrid E/M state of LECs, which was the specific mechanism of LN metastasis in the early stages of GC.


Asunto(s)
Anemia , Neoplasias Gástricas , Ratones , Animales , Humanos , Metástasis Linfática/patología , Neoplasias Gástricas/patología , Células Endoteliales/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Ratones Desnudos , Ganglios Linfáticos/patología , Anemia/patología
17.
Am J Vet Res ; 84(10): 1-6, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37586694

RESUMEN

OBJECTIVE: Compare erythropoiesis-related factors between different stages of canine chronic kidney disease (CKD). ANIMALS: 8 healthy adult dogs (controls), and 24 dogs with CKD, equally divided into 3 groups based on International Renal Interest Society-CKD Guidelines (stage 2, 3, and 4) were recruited between December 2012 and December 2014. METHODS: The following were assessed in all dogs and then compared between groups: bone marrow cytology, CBC, reticulocyte count, urinalysis, serum biochemistry, blood pressure, occult gastrointestinal bleeding, and serum concentrations of parathyroid hormone (PTH), erythropoietin, interleukin-1ß, interleukin-3, tumor necrosis factor-α (TNFα), and interferon-γ. RESULTS: Erythropoiesis inducing and suppressing factors and the results of the bone marrow cytology of dogs in stage 2 CKD did not differ from the control group. The presence of reticulocytosis in CKD stage 2 suggests that blood loss or erythrocyte destruction might be contributing to developing anemia. Anemia in dogs with progressive CKD was associated with increasing PTH and TNFα and with elevation of the ratio of myeloid to erythroid precursor cells caused by hypoplasia of the erythroid series. The latter was represented mainly by a decrease in the population of polychromatophilic rubricytes and metarubricytes. CLINICAL RELEVANCE: Increased PTH and TNFα seem to contribute to the reduced percentage of polychromatophilic rubricytes and erythroid population, thereby aggravating the anemia of dogs with advanced CKD. Gastrointestinal blood loss contributes to anemia in all canine CKD stages.


Asunto(s)
Anemia , Enfermedades de los Perros , Insuficiencia Renal Crónica , Perros , Animales , Células Precursoras Eritroides , Factor de Necrosis Tumoral alfa , Anemia/etiología , Anemia/veterinaria , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/veterinaria , Inflamación/complicaciones , Inflamación/veterinaria , Hemorragia Gastrointestinal/complicaciones , Hemorragia Gastrointestinal/veterinaria
18.
Front Immunol ; 14: 1202943, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545522

RESUMEN

Recent studies have demonstrated that a particular group of nucleated cells that exhibit erythroid markers (TER119 in mice and CD235a in humans) possess the ability to suppress the immune system and promote tumor growth. These cells are known as CD45+ erythroid progenitor cells (EPCs). According to our study, it appears that a subset of these CD45+ EPCs originate from B lymphocytes. Under conditions of hypoxia, mouse B lymphoma cells are capable of converting to erythroblast-like cells, which display phenotypes of CD45+TER119+ cells, including immunosuppressive effects on CD8 T cells. Furthermore, non-neoplastic B cells have similar differentiation abilities and exert the same immunosuppressive effect under anemia or tumor conditions in mice. Similar B cells exist in neonatal mice, which provides an explanation for the potential origin of immunosuppressive erythroid cells in newborns. Additionally, CD19+CD235a+ double-positive cells can be identified in the peripheral blood of patients with chronic lymphocytic leukemia. These findings indicate that some CD45+ EPCs are transdifferentiated from a selective population of CD19+ B lymphocytes in response to environmental stresses, highlighting the plasticity of B lymphocytes. We anticipate a potential therapeutic implication, in that targeting a specific set of B cells instead of erythroid cells should be expected to restore adaptive immunity and delay cancer progression.


Asunto(s)
Anemia , Eritroblastos , Humanos , Recién Nacido , Animales , Ratones , Eritroblastos/patología , Células Precursoras Eritroides , Diferenciación Celular , Linfocitos B/patología
19.
Rinsho Ketsueki ; 64(6): 482-488, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-37407471

RESUMEN

Large-scale in vitro red blood cell (RBC) production has been attempted in recent years. Potential cell sources for RBC production include hematopoietic stem/progenitor cells, pluripotent stem cells, and immortalized erythroid progenitor cell lines, which can induce enucleated RBCs with characteristics such as oxygen-carrying capacity and deformability. A phase I clinical study of cultured RBCs produced from hematopoietic stem/progenitor cells has revealed a similar in vivo half-life between cultured and native RBCs. Thus, the application of cultured RBCs in blood transfusion is gradually advancing. However, a single transfusion requires a large number of cells, unlike other cell therapies. Therefore, developing a method to mass-produce RBCs from a small culture volume at a low cost is important in the future. This review summarizes the current status and prospects concerning in vitro RBC production using each cell source, which can improve future transfusion medicine.


Asunto(s)
Medicina Transfusional , Humanos , Eritrocitos , Células Precursoras Eritroides/metabolismo , Eritropoyesis , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular
20.
Cells ; 12(8)2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37190030

RESUMEN

BACKGROUND: Sickle cell disease (SCD) is a highly prevalent genetic disease caused by a point mutation in the HBB gene, which can lead to chronic hemolytic anemia and vaso-occlusive events. Patient-derived induced pluripotent stem cells (iPSCs) hold promise for the development of novel predictive methods for screening drugs with anti-sickling activity. In this study, we evaluated and compared the efficiency of 2D and 3D erythroid differentiation protocols using a healthy control and SCD-iPSCs. METHODS: iPSCs were subjected to hematopoietic progenitor cell (HSPC) induction, erythroid progenitor cell induction, and terminal erythroid maturation. Differentiation efficiency was confirmed by flow cytometry analysis, colony-forming unit (CFU) assay, morphological analyses, and qPCR-based gene expression analyses of HBB and HBG2. RESULTS: Both 2D and 3D differentiation protocols led to the induction of CD34+/CD43+ HSPCs. The 3D protocol showed good efficiency (>50%) and high productivity (45-fold) for HSPC induction and increased the frequency of BFU-E, CFU-E, CFU-GM, and CFU-GEMM colonies. We also produced CD71+/CD235a+ cells (>65%) with a 630-fold cell expansion relative to that at the beginning of the 3D protocol. After erythroid maturation, we observed 95% CD235a+/DRAQ5- enucleated cells, orthochromatic erythroblasts, and increased expression of fetal HBG2 compared to adult HBB. CONCLUSION: A robust 3D protocol for erythroid differentiation was identified using SCD-iPSCs and comparative analyses; however, the maturation step remains challenging and requires further development.


Asunto(s)
Anemia de Células Falciformes , Células Madre Pluripotentes Inducidas , Adulto , Humanos , Diferenciación Celular , Células Madre Hematopoyéticas , Células Precursoras Eritroides/metabolismo , Anemia de Células Falciformes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA