Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 500
Filtrar
1.
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
2.
Adv Mater ; 34(19): e2200905, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35294781

RESUMEN

Due to the adverse effects of erythropoietin (EPO) on cancer patient survival, it is necessary to develop new agents that can be used to efficiently manage and treat cancer-related anemia. In this study, novel distinctive carbon dots, J-CDs, derived from jujube are designed, synthesized, and characterized. Based on the obtained results, this material comprises sp2 and sp3 carbon atoms, as well as oxygen/nitrogen-based groups, and it specifically promotes the proliferation of erythroid cells by stimulating the self-renewal of erythroid progenitor cells in vitro and in vivo. Moreover, J-CDs have no discernible effects on tumor proliferation and metastasis, unlike EPO. Transcriptome profiling suggests that J-CDs upregulate the molecules involved in hypoxia response, and they also significantly increase the phosphorylation levels of STAT5, the major transducer of signals for erythroid progenitor cell proliferation. Overall, this study demonstrates that J-CDs effectively promote erythrocyte production without affecting tumor proliferation and metastasis; thus, they may be promising agents for the treatment of cancer-related anemia.


Asunto(s)
Anemia , Eritropoyetina , Neoplasias , Anemia/tratamiento farmacológico , Anemia/patología , Carbono/farmacología , Carbono/uso terapéutico , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/fisiología , Eritropoyesis/fisiología , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Humanos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico
3.
Cancer Sci ; 113(2): 597-608, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34808021

RESUMEN

Several lines of research suggest that Bcl-xL-mediated anti-apoptotic effects may contribute to the pathogenesis of myeloproliferative neoplasms driven by JAK2V617F and serve as therapeutic target. Here, we used a knock-in JAK2V617F mouse model and confirmed that Bcl-xL was overexpressed in erythroid progenitors. The myeloproliferative neoplasm (MPN)-induced phenotype in the peripheral blood by conditional knock-in of JAK2V617F was abrogated by conditional knockout of Bcl2l1, which presented anemia and thrombocytopenia independently of JAK2 mutation status. Mx1-Cre Jak2V617W/VF /Bcl2l1f/f mice presented persistent splenomegaly as a result of extramedullary hematopoiesis and pro-apoptotic stimuli in terminally differentiated erythroid progenitors. The pan-BH3 mimetic inhibitor obatoclax showed superior cytotoxicity in JAK2V617F cell models, and reduced clonogenic capacity in ex vivo assay using Vav-Cre Jak2V617F bone marrow cells. Both ruxolitinib and obatoclax significantly reduced spleen weights in a murine Jak2V617F MPN model but did not show additive effect. The tumor burden reduction was observed with either ruxolitinib or obatoclax in terminal differentiation stage neoplastic cells but not in myeloid-erythroid precursors. Therefore, disrupting the BCL2 balance is not sufficient to treat MPN at the stem cell level, but it is certainly an additional option for controlling the critical myeloid expansion of the disease.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Janus Quinasa 2/antagonistas & inhibidores , Trastornos Mieloproliferativos/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Células Precursoras Eritroides/patología , Humanos , Indoles/uso terapéutico , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Nitrilos/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Carga Tumoral/efectos de los fármacos , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
4.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948226

RESUMEN

ß-thalassemias are among the most common inherited hemoglobinopathies worldwide and are the result of autosomal mutations in the gene encoding ß-globin, causing an absence or low-level production of adult hemoglobin (HbA). Induction of fetal hemoglobin (HbF) is considered to be of key importance for the development of therapeutic protocols for ß-thalassemia and novel HbF inducers need to be proposed for pre-clinical development. The main purpose on this study was to analyze Cinchona alkaloids (cinchonidine, quinidine and cinchonine) as natural HbF-inducing agents in human erythroid cells. The analytical methods employed were Reverse Transcription quantitative real-time PCR (RT-qPCR) (for quantification of γ-globin mRNA) and High Performance Liquid Chromatography (HPLC) (for analysis of the hemoglobin pattern). After an initial analysis using the K562 cell line as an experimental model system, showing induction of hemoglobin and γ-globin mRNA, we verified whether the two more active compounds, cinchonidine and quinidine, were able to induce HbF in erythroid progenitor cells isolated from ß-thalassemia patients. The data obtained demonstrate that cinchonidine and quinidine are potent inducers of γ-globin mRNA and HbF in erythroid progenitor cells isolated from nine ß-thalassemia patients. In addition, both compounds were found to synergize with the HbF inducer sirolimus for maximal production of HbF. The data obtained strongly indicate that these compounds deserve consideration in the development of pre-clinical approaches for therapeutic protocols of ß-thalassemia.


Asunto(s)
Alcaloides de Cinchona/farmacología , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biosíntesis , Talasemia beta/metabolismo , Células Precursoras Eritroides/patología , Humanos , Células K562 , Talasemia beta/tratamiento farmacológico
6.
Mayo Clin Proc ; 96(10): 2653-2659, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34489099

RESUMEN

The objective of this study is to describe the clinical features and outcomes of patients with the newly defined vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome. Nine men with somatic mutations in the UBA1 gene were identified; the most frequent variant was p.Met41Thr (7 of 9, 78%). The median age at VEXAS diagnosis was 74 (67, 76.5) years, and patients had a median duration of symptoms for 4 years before diagnosis. Refractory constitutional symptoms (88%), ear and nose chondritis (55%), and inflammatory arthritis (55%) were common clinical features. Vasculitis was noted in 44%. All patients had significantly elevated inflammatory markers and macrocytic anemia. Thrombocytopenia was present in 66% at diagnosis of VEXAS. Eight patients had bone marrow biopsies performed. All bone marrows were hypercellular, and there was vacuolization of the erythroid (100%) or myeloid precursors (75%). Glucocorticoids attenuated symptoms at prednisone doses ≥20 mg per day, but no other immunosuppressive agent showed consistent long-term control of disease. One patient with coexisting plasma-cell myeloma received plasma-cell-directed therapy with improvement of the inflammatory response, which is a novel finding. In conclusion, VEXAS syndrome is a clinically heterogeneous, treatment-refractory inflammatory condition caused by somatic mutation of the UBA1 gene. Patients often present with overlapping rheumatologic manifestations and persistent hematologic abnormalities. As such, internists and subspecialists, including pathologists, should be aware of this condition to avert diagnostic delay, now that the etiology of this syndrome is known.


Asunto(s)
Inflamación/diagnóstico , Síndromes Mielodisplásicos/diagnóstico , Enzimas Activadoras de Ubiquitina/genética , Anciano , Células Precursoras Eritroides/patología , Enfermedades Genéticas Congénitas , Enfermedades Genéticas Ligadas al Cromosoma X , Humanos , Inflamación/genética , Masculino , Mutación , Síndromes Mielodisplásicos/genética , Células Mieloides/patología , Vacuolas , Vasculitis/genética
8.
PLoS One ; 16(8): e0252156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34339420

RESUMEN

BACKGROUND: Kidney organoids have been broadly obtained from commercially available induced pluripotent stem cells (iPSCs); however, it has been a great challenge to efficiently produce renal organoid models from patients with autosomal dominant polycystic kidney disease (ADPKD) that recapitulate both embryogenesis and the mechanisms of cystogenesis. METHODS: Blood erythroid progenitors (EPs) from two ADPKD patients and one healthy donor (HC) was used as a comparative control to normalize the many technical steps for reprogramming EPs and for the organoids generation. EPs were reprogrammed by an episomal vector into iPSCs, which were differentiated into renal tubular organoids and then stimulated by forskolin to induce cysts formation. RESULTS: iPSCs derived from EPs exhibited all characteristics of pluripotency and were able to differentiate into all three germ layers. 3D tubular organoids were generated from single cells after 28 days in Matrigel. HC and ADPKD organoids did not spontaneously form cysts, but upon forskolin stimulation, cysts-like structures were observed in the ADPKD organoids but not in the HC-derived organoids. CONCLUSION: The findings of this study showed that kidney organoids were successfully generated from the blood EP cells of ADPKD patients and a healthy control donor. This approach should contribute as a powerful tool for embryonic kidney development model, which is able to recapitulate the very early pathophysiological mechanisms involved in cytogenesis.


Asunto(s)
Células Precursoras Eritroides , Células Madre Pluripotentes Inducidas , Riñón , Organoides , Riñón Poliquístico Autosómico Dominante , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Riñón/metabolismo , Riñón/patología , Organoides/metabolismo , Organoides/patología , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología
9.
Medicine (Baltimore) ; 100(15): e25528, 2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33847676

RESUMEN

RATIONALE: Pure erythroid leukemia is a rare subcategory of acute myeloid leukemia characterized by predominant immature erythroid population. Its occurrence subsequent to acute myelomonocytic leukemia has not been reported before. We reported this rare case to call attention because it may pose a diagnostic challenge. PATIENTS CONCERNS: A 54-year-old female patient presented to our hospital in March 2018 with symptoms of easy fatigability. DIAGNOSIS: Bone marrow aspiration was hypercellular showing 67.2% blasts mainly including moderate myeloblasts and monoblasts. There was mild dysplasia with some cells having round, oval, or bizarre nuclei which containing 1 to 3 nucleolus. Erythroid lineage was hypoplasia and mature erythrocytes were generally normal. Conventional cytogenetics of bone marrow cells revealed complex karyotype (44, XX, del (5) (q14q34) del (5) (q14q34), del (14) t (11;14) (q10; q10), -16, del (17), -18[10]). INTERVENTIONS: The patient was treated with second line chemotherapy but did not respond. QUTCOMES: She died of cardiopulmonary failure 19days after starting of therapy. LESSONS: This unexpected and relatively uncommon occurrence was associated with a universally rapid and fatal clinical course with survival measured in <2 months despite intensive chemotherapy. We call attention to this rare phenomenon because it may pose a diagnostic challenge.


Asunto(s)
Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Mielomonocítica Aguda/diagnóstico , Células de la Médula Ósea/patología , Análisis Citogenético , Diagnóstico Diferencial , Células Precursoras Eritroides/patología , Resultado Fatal , Femenino , Humanos , Cariotipificación , Leucemia Eritroblástica Aguda/etiología , Leucemia Mielomonocítica Aguda/complicaciones , Ilustración Médica , Persona de Mediana Edad
10.
Exp Hematol ; 97: 66-78.e6, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33631277

RESUMEN

Rpl11 haploinsufficient mice develop a macrocytic anemia similar to patients with DBA. Here, we fully characterize this model from clinical and pathophysiological perspectives. Early erythroid precursors have increased heme content and high cytoplasmic reactive oxygen species, impairing erythroid differentiation at the colony-forming unit-erythroid (CFU-E)/proerythroblast stage and subsequently. Using single-cell analyses that link a cell's surface protein expression to its total transcriptome and unbiased analyses, we found GATA1, GATA1 target gene, and mitotic spindle pathway gene transcription were the pathways that decreased the most. Expression of ribosome protein and globin genes was amplified. These changes, as well as the other transcriptional changes that were identified, closely resemble findings in mice that lack the heme export protein FLVCR and, thus, suggest that heme excess and toxicity are the primary drivers of the macrocytic anemia. Consistent with this, treating Rpl11 haploinsufficient mice with corticosteroids increased the numbers of earliest erythroblasts but failed to overcome heme toxicity and improve the anemia. Rpl11 haploinsufficient mice uniquely upregulated mitochondrial genes, p53 and CDKN1A pathway genes, and DNA damage checkpoint genes, which should contribute further to erythroid marrow failure. Together our data establish Rpl11 haploinsufficient mice as an excellent model of DBA that can be used to study DBA pathogenesis and test novel therapies.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Eritropoyesis , Haploinsuficiencia , Anemia de Diamond-Blackfan/patología , Animales , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Femenino , Humanos , Masculino , Ratones , Análisis de la Célula Individual
11.
Blood Cells Mol Dis ; 87: 102522, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33260083

RESUMEN

Stathmin 1 (STMN1) is a cytosolic phosphoprotein that was discovered as a result of its high level of expression in leukemic cells. It plays an important role in the regulation of mitosis by promoting depolymerization of the microtubules that make up the mitotic spindle and, aging has been shown to impair STMN1 levels and change microtubule stability. We have previously demonstrated that a high level of STMN1 expression during early megakaryopoiesis is necessary for proliferation of megakaryocyte progenitors and that down-regulation of STMN1 expression during late megakaryopoiesis is important for megakaryocyte maturation and platelet production. In this report, we examined the effects of STMN1 deficiency on erythroid and megakaryocytic lineages in the mouse. Our studies show that STMN1 deficiency results in mild thrombocytopenia in young animals which converts into profound thrombocythemia as the mice age. STMN1 deficiency also lead to macrocytic changes in both erythrocytes and megakaryocytes that persisted throughout the life of STMN1 knock-out mice. Furthermore, STMN1 knock-out mice displayed a lower number of erythroid and megakaryocytic progenitor cells and had delayed recovery of their blood counts after chemotherapy. These studies show an important role for STMN1 in normal erythro-megakaryopoietic development and suggests potential implications for disorders affecting these hematopoietic lineages.


Asunto(s)
Anemia Macrocítica/genética , Células Precursoras Eritroides/patología , Megacariocitos/patología , Estatmina/genética , Trombocitosis/genética , Anemia Macrocítica/patología , Animales , Plaquetas/patología , Eritropoyesis , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitosis/patología
12.
Front Immunol ; 11: 597433, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329589

RESUMEN

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Asunto(s)
Antígenos CD/inmunología , Células Precursoras Eritroides , Terapia de Inmunosupresión , Listeria monocytogenes/inmunología , Listeriosis , Receptores de Transferrina/inmunología , Animales , Animales Recién Nacidos , COVID-19/inmunología , COVID-19/patología , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/trasplante , Femenino , Xenoinjertos , Humanos , Recién Nacido , Listeriosis/inmunología , Listeriosis/patología , Listeriosis/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2/inmunología
13.
Exp Cell Res ; 397(2): 112346, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33164866

RESUMEN

Growth differentiation factor-15 (GDF-15) is a member of TGF-ß superfamily. Among hematopoietic cells, this factor is mainly produced by erythroid series and is recently considered a biomarker of ineffective erythropoiesis (IE). Whether IE induces enhanced GDF-15 expression or is prompted by it, has remained elusive. In this study we investigated how high levels of GDF-15 contribute to IE-associated erythroid dysplasia. We assessed mRNA levels of GDF-15 during erythroid maturation as well as in patients with IE using qRT-PCR. Later, the erythroid colony-forming capacity of GDF-15-treated hematopoietic stem cells (HSCs) was evaluated by CFC assay. Any effect of elevated levels of GDF-15 on erythroid maturation was ultimately examined by expression analysis of erythroid-associated transcription factors and flow cytometry analysis of CD235a expression. GDF-15 mRNA expression increased during erythroid differentiation and also in ß-thalassemia and MDS patients which was directly correlated with erythropoiesis severity. Treating the cells with high GDF-15 concentration (50 ng/ml) resulted in an approximate 30% decline in the capacity of erythroid colony formation of HSCs and CD235a positive cells. Additionally, erythroid-specific transcription factors showed significant down-regulation in the early stages of erythroid differentiation. According to the expression level of GDF-15 and the role it plays in the erythroid system, high-levels of this factor could be an auto-modulatory mechanism to control the excessive production of erythroid cells.


Asunto(s)
Células Precursoras Eritroides/patología , Eritropoyesis , Factor 15 de Diferenciación de Crecimiento/metabolismo , Células Madre Hematopoyéticas/patología , Hiperplasia/patología , Talasemia beta/patología , Estudios de Casos y Controles , Diferenciación Celular , Células Precursoras Eritroides/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Hiperplasia/metabolismo , Factor de Células Madre/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Talasemia beta/metabolismo
14.
Sci Rep ; 10(1): 18469, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33116141

RESUMEN

Bone marrow erythropoiesis is mainly homeostatic and a demand of oxygen in tissues activates stress erythropoiesis in the spleen. Here, we show an increase in the number of circulating erythrocytes in apolipoprotein E-/- mice fed a Western high-fat diet, with similar number of circulating leukocytes and CD41+ events (platelets). Atherogenic conditions increase spleen erythropoiesis with no variations of this cell lineage in the bone marrow. Spleens from atherogenic mice show augmented number of late-stage erythroblasts and biased differentiation of progenitor cells towards the erythroid cell lineage, with an increase of CD71+CD41CD34-CD117+Sca1-Lin- cells (erythroid-primed megakaryocyte-erythroid progenitors), which is consistent with the way in which atherogenesis modifies the expression of pro-erythroid and pro-megakaryocytic genes in megakaryocyte-erythroid progenitors. These data explain the transiently improved response to an acute severe hemolytic anemia insult found in atherogenic mice in comparison to control mice, as well as the higher burst-forming unit-erythroid and colony forming unit-erythroid capacity of splenocytes from atherogenic mice. In conclusion, our work demonstrates that, along with the well stablished enhancement of monocytosis during atherogenesis, stress erythropoiesis in apolipoprotein E-/- mice fed a Western high fat diet results in increased numbers of circulating red blood cells.


Asunto(s)
Aterosclerosis/metabolismo , Diferenciación Celular , Células Precursoras Eritroides/metabolismo , Eritropoyesis , Hematopoyesis Extramedular , Bazo/metabolismo , Estrés Fisiológico , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Células Precursoras Eritroides/patología , Ratones , Ratones Noqueados para ApoE , Bazo/patología
15.
J Exp Med ; 217(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32520308

RESUMEN

An important comorbidity of chronic inflammation is anemia, which may be related to dysregulated activity of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Among HSPCs, we found that the receptor for IL-33, ST2, is expressed preferentially and highly on erythroid progenitors. Induction of inflammatory spondyloarthritis in mice increased IL-33 in BM plasma, and IL-33 was required for inflammation-dependent suppression of erythropoiesis in BM. Conversely, administration of IL-33 in healthy mice suppressed erythropoiesis, decreased hemoglobin expression, and caused anemia. Using purified erythroid progenitors in vitro, we show that IL-33 directly inhibited terminal maturation. This effect was dependent on NF-κB activation and associated with altered signaling events downstream of the erythropoietin receptor. Accordingly, IL-33 also suppressed erythropoietin-accelerated erythropoiesis in vivo. These results reveal a role for IL-33 in pathogenesis of anemia during inflammatory disease and define a new target for its treatment.


Asunto(s)
Anemia/patología , Diferenciación Celular , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Inflamación/patología , Interleucina-33/metabolismo , Anemia/complicaciones , Animales , Anexina A5/metabolismo , Médula Ósea/patología , Enfermedad Crónica , Eritropoyesis , Eritropoyetina/farmacología , Hematopoyesis , Inflamación/complicaciones , Inyecciones , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Antígeno Ki-67/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Biológicos , Mielopoyesis , FN-kappa B/metabolismo , Fosforilación , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Espondiloartritis/patología , beta-Glucanos
18.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32421753

RESUMEN

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Asunto(s)
Células Precursoras Eritroides/inmunología , Eritropoyesis/inmunología , Mastocitos/inmunología , Mastocitosis/inmunología , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Animales , Anhidrasa Carbónica I/genética , Anhidrasa Carbónica I/inmunología , Células Precursoras Eritroides/parasitología , Células Precursoras Eritroides/patología , Femenino , Mastocitos/parasitología , Mastocitos/patología , Mastocitosis/genética , Mastocitosis/patología , Ratones , Ratones Transgénicos , Triquinelosis/genética , Triquinelosis/patología
19.
Cancer Cell ; 37(5): 690-704.e8, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32330454

RESUMEN

Acute erythroid leukemia (AEL) commonly involves both myeloid and erythroid lineage transformation. However, the mutations that cause AEL and the cell(s) that sustain the bilineage leukemia phenotype remain unknown. We here show that combined biallelic Cebpa and Gata2 zinc finger-1 (ZnF1) mutations cooperatively induce bilineage AEL, and that the major leukemia-initiating cell (LIC) population has a neutrophil-monocyte progenitor (NMP) phenotype. In pre-leukemic NMPs Cebpa and Gata2 mutations synergize by increasing erythroid transcription factor (TF) expression and erythroid TF chromatin access, respectively, thereby installing ectopic erythroid potential. This erythroid-permissive chromatin conformation is retained in bilineage LICs. These results demonstrate that synergistic transcriptional and epigenetic reprogramming by leukemia-initiating mutations can generate neomorphic pre-leukemic progenitors, defining the lineage identity of the resulting leukemia.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Linaje de la Célula , Transformación Celular Neoplásica/patología , Células Precursoras Eritroides/patología , Factor de Transcripción GATA2/genética , Leucemia Eritroblástica Aguda/patología , Mutación , Neutrófilos/patología , Anciano , Alelos , Animales , Diferenciación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Femenino , Factor de Transcripción GATA1/genética , Humanos , Leucemia Eritroblástica Aguda/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neutrófilos/metabolismo , Dedos de Zinc
20.
J Clin Invest ; 130(4): 2097-2110, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961825

RESUMEN

Despite the effective clinical use of steroids for the treatment of Diamond Blackfan anemia (DBA), the mechanisms through which glucocorticoids regulate human erythropoiesis remain poorly understood. We report that the sensitivity of erythroid differentiation to dexamethasone is dependent on the developmental origin of human CD34+ progenitor cells, specifically increasing the expansion of CD34+ progenitors from peripheral blood (PB) but not cord blood (CB). Dexamethasone treatment of erythroid-differentiated PB, but not CB, CD34+ progenitors resulted in the expansion of a newly defined CD34+CD36+CD71hiCD105med immature colony-forming unit-erythroid (CFU-E) population. Furthermore, proteomics analyses revealed the induction of distinct proteins in dexamethasone-treated PB and CB erythroid progenitors. Dexamethasone treatment of PB progenitors resulted in the specific upregulation of p57Kip2, a Cip/Kip cyclin-dependent kinase inhibitor, and we identified this induction as critical; shRNA-mediated downregulation of p57Kip2, but not the related p27Kip1, significantly attenuated the impact of dexamethasone on erythroid differentiation and inhibited the expansion of the immature CFU-E subset. Notably, in the context of DBA, we found that steroid resistance was associated with dysregulated p57Kip2 expression. Altogether, these data identify a unique glucocorticoid-responsive human erythroid progenitor and provide new insights into glucocorticoid-based therapeutic strategies for the treatment of patients with DBA.


Asunto(s)
Anemia de Diamond-Blackfan/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Dexametasona/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adulto , Anemia de Diamond-Blackfan/tratamiento farmacológico , Anemia de Diamond-Blackfan/patología , Antígenos CD/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Células Precursoras Eritroides/patología , Femenino , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...