Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Front Immunol ; 15: 1381919, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799424

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos , Células Precursoras Eritroides , Tolerância Imunológica , Animais , Linfócitos T CD8-Positivos/imunologia , Camundongos , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos Endogâmicos C57BL , Transferência Adotiva , Espécies Reativas de Oxigênio/metabolismo , Evasão Tumoral/imunologia , Linhagem Celular Tumoral , Ácido Úrico
2.
Exp Hematol ; 89: 43-54.e2, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32750404

RESUMO

Steady-state erythropoiesis generates new erythrocytes at a constant rate, and it has enormous productive capacity. This production is balanced by the removal of senescent erythrocytes by macrophages in the spleen and liver. Erythroid homeostasis is highly regulated to maintain sufficient erythrocytes for efficient oxygen delivery to the tissues, while avoiding viscosity problems associated with overproduction. However, there are times when this constant production of erythrocytes is inhibited or is inadequate; at these times, erythroid output is increased to compensate for the loss of production. In some cases, increased steady-state erythropoiesis can offset the loss of erythrocytes but, in response to inflammation caused by infection or tissue damage, steady-state erythropoiesis is inhibited. To maintain homeostasis under these conditions, an alternative stress erythropoiesis pathway is activated. Emerging data suggest that the bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis pathway is integrated into the inflammatory response and generates a bolus of new erythrocytes that maintain homeostasis until steady-state erythropoiesis can resume. In this perspective, we define the mechanisms that generate new erythrocytes when steady-state erythropoiesis is impaired and discuss experimental models to study human stress erythropoiesis.


Assuntos
Proteína Morfogenética Óssea 4/genética , Eritrócitos/citologia , Células Precursoras Eritroides/citologia , Eritropoese/genética , Macrófagos/citologia , Estresse Fisiológico/genética , Animais , Proteína Morfogenética Óssea 4/imunologia , Senescência Celular/imunologia , Citocinas/genética , Citocinas/imunologia , Eritrócitos/imunologia , Células Precursoras Eritroides/imunologia , Eritropoese/imunologia , Regulação da Expressão Gênica , Humanos , Inflamação , Fígado/citologia , Fígado/imunologia , Macrófagos/imunologia , Camundongos , Modelos Biológicos , Fagocitose , Baço/citologia , Baço/imunologia , Estresse Fisiológico/imunologia
3.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32421753

RESUMO

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.


Assuntos
Células Precursoras Eritroides/imunologia , Eritropoese/imunologia , Mastócitos/imunologia , Mastocitose/imunologia , Trichinella spiralis/imunologia , Triquinelose/imunologia , Animais , Anidrase Carbônica I/genética , Anidrase Carbônica I/imunologia , Células Precursoras Eritroides/parasitologia , Células Precursoras Eritroides/patologia , Feminino , Mastócitos/parasitologia , Mastócitos/patologia , Mastocitose/genética , Mastocitose/patologia , Camundongos , Camundongos Transgênicos , Triquinelose/genética , Triquinelose/patologia
4.
FEBS Open Bio ; 10(3): 427-433, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31977161

RESUMO

Renal anemia in chronic kidney disease is treated with recombinant human erythropoietin (rhEPO). However, some patients with anemia do not respond well to rhEPO, emphasizing the need for a more biocompatible EPO. Differentiation protocols for hepatic lineages have been modified to enable production from human induced pluripotent stem cell (hiPSC)-derived EPO-producing cells (EPO cells). However, markers for hiPSC-EPO cells are lacking, making it difficult to purify hiPSC-EPO cells and therefore to optimize EPO production and cell counts for transplantation. To address these issues, we investigated whether CD140b and CD73 could be used as markers for hiPSC-EPO cells. We measured the expression of EPO, CD140b, and CD73 in hiPSC-EPO cells and the EPO concentration in the cell supernatant by immunohistochemistry and enzyme-linked immunosorbent assays on culture day 13, revealing that expression levels of CD140b and CD73 are correlated with the level of EPO. In addition, rates of CD140b+ CD73+ cells were observed to be correlated with the concentration of EPO. Thus, our results suggest that CD140b and CD73 may be markers for hiPSC-EPO cells.


Assuntos
5'-Nucleotidase/imunologia , Células Precursoras Eritroides/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/imunologia , 5'-Nucleotidase/metabolismo , Anemia/sangue , Anemia/metabolismo , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Células Precursoras Eritroides/imunologia , Eritropoetina/metabolismo , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/metabolismo
5.
Sci Signal ; 12(598)2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506384

RESUMO

Inflammation alters bone marrow hematopoiesis to favor the production of innate immune effector cells at the expense of lymphoid cells and erythrocytes. Furthermore, proinflammatory cytokines inhibit steady-state erythropoiesis, which leads to the development of anemia in diseases with chronic inflammation. Acute anemia or hypoxic stress induces stress erythropoiesis, which generates a wave of new erythrocytes to maintain erythroid homeostasis until steady-state erythropoiesis can resume. Although hypoxia-dependent signaling is a key component of stress erythropoiesis, we found that inflammation also induced stress erythropoiesis in the absence of hypoxia. Using a mouse model of sterile inflammation, we demonstrated that signaling through Toll-like receptors (TLRs) paradoxically increased the phagocytosis of erythrocytes (erythrophagocytosis) by macrophages in the spleen, which enabled expression of the heme-responsive gene encoding the transcription factor SPI-C. Increased amounts of SPI-C coupled with TLR signaling promoted the expression of Gdf15 and Bmp4, both of which encode ligands that initiate the expansion of stress erythroid progenitors (SEPs) in the spleen. Furthermore, despite their inhibition of steady-state erythropoiesis in the bone marrow, the proinflammatory cytokines TNF-α and IL-1ß promoted the expansion and differentiation of SEPs in the spleen. These data suggest that inflammatory signals induce stress erythropoiesis to maintain erythroid homeostasis when inflammation inhibits steady-state erythropoiesis.


Assuntos
Proteínas de Ligação a DNA/imunologia , Eritropoese/imunologia , Heme/imunologia , Inflamação/imunologia , Estresse Fisiológico/imunologia , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/imunologia , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Eritrócitos/imunologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/metabolismo , Eritropoese/genética , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/imunologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Heme/metabolismo , Inflamação/genética , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Baço/imunologia , Baço/metabolismo , Estresse Fisiológico/genética
6.
Nat Med ; 24(10): 1536-1544, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30297899

RESUMO

Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.


Assuntos
Anemia/imunologia , Células Precursoras Eritroides/imunologia , Células Supressoras Mieloides/imunologia , Sarcoma Mieloide/imunologia , Anemia/genética , Anemia/patologia , Animais , Antígenos CD/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Humanos , Tolerância Imunológica , Imunidade Inata/genética , Antígenos Comuns de Leucócito/imunologia , Camundongos , Estadiamento de Neoplasias , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/imunologia , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patologia , Linfócitos T Reguladores/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nat Commun ; 9(1): 1828, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739946

RESUMO

NOTCH signaling is required for the arterial specification and formation of hematopoietic stem cells (HSCs) and lympho-myeloid progenitors in the embryonic aorta-gonad-mesonephros region and extraembryonic vasculature from a distinct lineage of vascular endothelial cells with hemogenic potential. However, the role of NOTCH signaling in hemogenic endothelium (HE) specification from human pluripotent stem cell (hPSC) has not been studied. Here, using a chemically defined hPSC differentiation system combined with the use of DLL1-Fc and DAPT to manipulate NOTCH, we discover that NOTCH activation in hPSC-derived immature HE progenitors leads to formation of CD144+CD43-CD73-DLL4+Runx1 + 23-GFP+ arterial-type HE, which requires NOTCH signaling to undergo endothelial-to-hematopoietic transition and produce definitive lympho-myeloid and erythroid cells. These findings demonstrate that NOTCH-mediated arterialization of HE is an essential prerequisite for establishing definitive lympho-myeloid program and suggest that exploring molecular pathways that lead to arterial specification may aid in vitro approaches to enhance definitive hematopoiesis from hPSCs.


Assuntos
Artérias/citologia , Endotélio Vascular/citologia , Hemangioblastos/citologia , Hematopoese , Neovascularização Fisiológica , Células-Tronco Pluripotentes/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Antígenos CD/imunologia , Artérias/metabolismo , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Rastreamento de Células/instrumentação , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Endotélio Vascular/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/imunologia , Hemangioblastos/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células-Tronco Pluripotentes/imunologia
8.
Clin Lab ; 63(2): 373-378, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28182355

RESUMO

BACKGROUND: Pure red cell aplasia (PRCA) is an uncommon disease which involves an almost complete absence of the erythroid lineage in bone marrow (BM) and causes severe anemia. Cases due to monoclonal gammopathy occurring in plasma cell disorder have been infrequently reported. Here we report a case of PRCA associated plasma cell disorder, especially monoclonal gammopathy of undetermined significance (MGUS). METHODS: A 55-year-old male visited the ER due to general weakness. At his initial visit he exhibited severe anemia. Mild intravascular hemolysis was suspected. For anemia evaluation, BM examination was performed. In BM aspiration, almost no erythroid precursor cells were observed. Also, plasma cells were relatively elevated, at 7.2%. Serum electrophoresis and immunofixation revealed paraproteinemia of 5.1 g/L (IgG and lambda). No hypercalcemia, renal insufficiency or lytic bone lesions were found. This unusual case showed MGUS accompanied by PCRA. We were also able to assume the erythroid cell-specific restriction due to paraprotein, because we ruled out possible causes of PRCA. RESULTS: We discovered several reported cases associated with plasma cell dyscrasia. However, most of these cases involved plasma cell myeloma, characterized by high immunoglobulin burden. Our case demonstrates that PRCA is also observed in cases with MGUS, where immunoglobulin burden is low. CONCLUSIONS: It is not yet accurately known, what parts of erythroid precursors are targeted by M-protein nor what the mechanism is. Therefore, additional research into this matter is necessary.


Assuntos
Gamopatia Monoclonal de Significância Indeterminada/complicações , Mieloma Múltiplo/complicações , Aplasia Pura de Série Vermelha/complicações , Anemia/etiologia , Biomarcadores/sangue , Exame de Medula Óssea , Transfusão de Eritrócitos , Células Precursoras Eritroides/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/imunologia , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/imunologia , Proteínas do Mieloma/imunologia , Aplasia Pura de Série Vermelha/diagnóstico , Aplasia Pura de Série Vermelha/imunologia , Aplasia Pura de Série Vermelha/terapia
9.
J Clin Pathol ; 69(10): 933-7, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27484914

RESUMO

The reaction of Hector Battifora mesothelial epitope-1 (HBME-1) antibody with scattered pronormoblasts in normal bone marrow core biopsy specimens has been reported. This study evaluated the immunohistochemical profile of HBME-1 in a panel of 52 normal, dyserythropoietic and neoplastic marrow samples. We compared the staining property of HBME-1 with that of the commonly used erythroid marker, glycophorin A (CD235a) and in each case, we semi-quantitatively evaluated the HBME-1/CD235a-positive cells ratio. In normal samples, HBME-1 labelled scattered immature erythroid precursors. In dyserythropoietic specimens, HBME-1 stained nucleated erythroid precursors in varying degrees, from pronormoblast through normoblast stages, with the highest intensity in immature forms. Overall, the cellular background of non-erythroid progenitors, erythrocytes and neoplastic cells did not react with HBME-1, except in leukaemia cases with myelodysplasia-related changes. Our study shows that HBME-1 is a useful marker to identify immature erythroid precursors and that an HBME-1/CD235a-positive cells ratio ≥10% is associated with dyserythropoiesis.


Assuntos
Biomarcadores Tumorais/metabolismo , Medula Óssea/patologia , Células Precursoras Eritroides/metabolismo , Síndromes Mielodisplásicas/metabolismo , Anticorpos Monoclonais/imunologia , Biomarcadores Tumorais/imunologia , Biópsia com Agulha de Grande Calibre , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/patologia , Glicoforinas/metabolismo , Humanos , Síndromes Mielodisplásicas/diagnóstico , Estudos Retrospectivos
10.
J Pediatr Hematol Oncol ; 37(6): e375-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25929611

RESUMO

Hemolytic disease of the newborn (HDN) arising from MNSs incompatibility is rare, with few reports of prolonged anemia and reticulocytopenia following HDN. We report the younger of 2 male siblings, both of whom had anti-M-induced HDN and anemia persisting for over a month. Peripheral reticulocytes remained inappropriately low for the degree of anemia, and they needed multiple red cell transfusions. Viral infections were ruled out. Corticosteroids were given for suspected pure red cell aplasia. Anemia and reticulocytopenia subsequently improved. Colony-forming unit erythroid assay revealed erythropoietic suppression of M antigen-positive erythroid precursor cells cultured with maternal or infant sera containing anti-M. In conclusion, maternal anti-M caused HDN and prolonged anemia by erythropoietic suppression in 2 siblings.


Assuntos
Anemia/etiologia , Eritroblastose Fetal/etiologia , Células Precursoras Eritroides/patologia , Eritropoese/imunologia , Imunoglobulina M/imunologia , Isoanticorpos/imunologia , Aplasia Pura de Série Vermelha/complicações , Adulto , Anemia/patologia , Diferenciação Celular , Ensaio de Unidades Formadoras de Colônias , Eritroblastose Fetal/patologia , Células Precursoras Eritroides/imunologia , Feminino , Humanos , Imunoglobulina M/sangue , Lactente , Recém-Nascido , Masculino , Gravidez , Prognóstico , Irmãos
11.
Exp Mol Pathol ; 95(3): 364-75, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24184702

RESUMO

We performed a series of experiments under a working hypothesis that cross-linked oligomers of ribosomal protein S19 (RP S19) play an essential role in definitive erythropoiesis as a ligand of the C5a receptor of erythroblasts and macrophages. We found molecules functionally and immunologically indistinguishable from RP S19 oligomers in the extracellular fluid of porcine and guinea pig bone marrow. When an increased hematopoietic state was induced in guinea pigs by bloodletting, the bone marrow RP S19 oligomer concentration was concomitantly increased. However, when the RP S19 oligomers were immunologically neutralized or the C5a receptor was pharmacologically antagonized, hyper-erythropoiesis induced by bloodletting was prevented and the anemic state was retarded in guinea pigs. When the RP S19 oligomers were neutralized in mice after bloodletting, the reactive hyper proliferation of erythroblasts in the spleen was prevented. Proerythroblasts and erythroblasts prepared by bone marrow aspiration from healthy individuals were found to express significant levels of the C5a receptor and type 2 transglutaminase genes. Majority of erythroblasts in cord blood of healthy newborns bore the C5a receptor. Taken together, these results support our hypothesis.


Assuntos
Medula Óssea/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Receptor da Anafilatoxina C5a/metabolismo , Proteínas Ribossômicas/metabolismo , Adulto , Animais , Anticorpos Neutralizantes , Sangria , Western Blotting , Medula Óssea/imunologia , Medula Óssea/patologia , Reagentes de Ligações Cruzadas , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/patologia , Líquido Extracelular/imunologia , Líquido Extracelular/metabolismo , Feminino , Sangue Fetal/imunologia , Sangue Fetal/metabolismo , Citometria de Fluxo , Proteínas de Ligação ao GTP/metabolismo , Cobaias , Hemoglobinas/metabolismo , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Proteína 2 Glutamina gama-Glutamiltransferase , Multimerização Proteica , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Proteínas Ribossômicas/química , Proteínas Ribossômicas/imunologia , Suínos , Transglutaminases/metabolismo
12.
Blood Rev ; 27(1): 55-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23261066

RESUMO

Donor lymphocyte infusion (DLI) using unstimulated leukapheresis is one of the most effective treatment strategies for patients with hematological malignancies; its graft-versus-leukemia effects make it especially effective in chronic myeloid leukemia patients who relapsed after allogeneic stem cell transplantation (allo-HSCT). However, DLI application is limited by the development of graft-versus-host disease and aplasia, and thus cannot be routinely applied for prophylaxis. Therefore, important questions remain to be answered, such as when, and whom to DLI? Recent advances enable DLI using allografts of granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells; allodepleted donor T cells; and infusions of donor-derived, ex vivo-expanded, CD8(+) cytotoxic T lymphocyte, which can decrease relapse and improve transplant outcomes. Preemptive immunotherapy of relapse was also introduced based on the determination of mixed chimerism and minimal residual disease. In this review, we summarize the latest developments in recent strategies that will affect future DLI efficacy - focusing on the disadvantages and advantages of each protocol for the treatment, preemptive therapy, and prophylaxis of relapse.


Assuntos
Células Precursoras Eritroides/transplante , Doença Enxerto-Hospedeiro/terapia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Imunização Passiva , Transfusão de Linfócitos , Linfócitos T/transplante , Quimerismo , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/mortalidade , Doença Enxerto-Hospedeiro/patologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Mobilização de Células-Tronco Hematopoéticas , Humanos , Seleção de Pacientes , Recidiva , Análise de Sobrevida , Linfócitos T/imunologia , Transplante Homólogo
13.
Br J Haematol ; 160(2): 244-50, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23151030

RESUMO

The prevalence, clinical associations and pathogenic role of newly identified autoantibodies to the erythropoietin receptor (EPOR) in patients with anaemia were investigated. Sera from 203 patients with immune-related or chronic kidney diseases were screened for anti-EPOR antibodies by enzyme-linked immunosorbent assay, and antibody specificity was evaluated by immunoprecipitating EPOR from AS-E2 cells using purified immunoglobulin (Ig) fractions. In addition, the pathogenic role of anti-EPOR antibodies was determined by examining their inhibitory effects on AS-E2 cell proliferation. Clinical findings were compared between patients with and without anti-EPOR antibodies, in all patients and those with systemic lupus erythematosus (SLE). Serum anti-EPOR antibodies were detected in 52 patients. Purified IgG or IgM fractions from anti-EPOR antibody-positive sera immunoprecipitated EPOR and inhibited the EPO-dependent proliferation of AS-E2 cells in a dose-dependent manner. Anti-EPOR antibodies were associated with low haemoglobin concentrations and reticulocytopenia in all patients enrolled and those with SLE. Further, there was a negative correlation between the levels of anti-EPOR antibodies and the number of bone marrow erythroblasts in patients who underwent bone marrow examinations. These findings suggest that EPOR autoantibodies are present in a subset of patients with anaemia and that impaired erythropoiesis can be mediated by anti-EPOR antibodies, which functionally neutralize EPO activity.


Assuntos
Anemia/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Eritropoese/imunologia , Nefropatias/imunologia , Receptores da Eritropoetina/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Especificidade de Anticorpos , Autoanticorpos/sangue , Doenças Autoimunes/sangue , Medula Óssea/patologia , Linhagem Celular/efeitos dos fármacos , Criança , Doença Crônica , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/patologia , Feminino , Hemoglobinas/análise , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Nefropatias/sangue , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Transtornos Linfoproliferativos/sangue , Transtornos Linfoproliferativos/imunologia , Masculino , Pessoa de Meia-Idade , Contagem de Reticulócitos , Adulto Jovem
14.
J Virol ; 85(11): 5423-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21411527

RESUMO

Natural killer (NK) cells function as early effector cells in the innate immune defense against viral infections and also participate in the regulation of normal and malignant hematopoiesis. NK cell activities have been associated with early clearance of viremia in experimental simian immunodeficiency virus and clinical human immunodeficiency virus type 1 (HIV-1) infections. We have previously shown that NK cells function as major cytotoxic effector cells in vaccine-induced immune protection against Friend virus (FV)-induced leukemia, and NK cell depletion totally abrogates the above protective immunity. However, how NK cells recognize retrovirus-infected cells remains largely unclear. The present study demonstrates a correlation between the expression of the products of retinoic acid early transcript-1 (RAE-1) genes in target cells and their susceptibility to killing by NK cells isolated from FV-infected animals. This killing was abrogated by antibodies blocking the NKG2D receptor in vitro. Further, the expression of RAE-1 proteins on erythroblast surfaces increased early after FV inoculation, and administration of an RAE-1-blocking antibody resulted in increased spleen infectious centers and exaggerated pathology, indicating that FV-infected erythroid cells are recognized by NK cells mainly through the NKG2D-RAE-1 interactions in vivo. Enhanced retroviral replication due to host gene-targeting resulted in markedly increased RAE-1 expression in the absence of massive erythroid cell proliferation, indicating a direct role of retroviral replication in RAE-1 upregulation.


Assuntos
Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/virologia , Vírus da Leucemia Murina de Friend/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
15.
Bone Marrow Transplant ; 46(5): 650-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20711241

RESUMO

The CD34(+) compartment of grafts for clinical allogeneic hematopoietic cell transplantation (HCT) is very heterogeneous. It contains hematopoietic stem cells and several different progenitor cell populations. This study assesses (1) the content of these populations in clinical grafts from G-CSF-mobilized PBMCs, BM and cord blood, (2) the functional correlation of the graft composition with time to engraftment of neutrophils, platelets and reticulocytes and (3) donor age-related changes. Quantitative flow cytometry showed that the distribution of the progenitor subsets differed significantly between the graft sources and that donor age-related changes occur. In patients after myeloablative allogeneic HCT, accelerated platelet and reticulocyte engraftment correlated with the content of common myeloid and/or megakaryocyte erythroid progenitors in the graft. These findings show that a better understanding of the progenitor compartment in human hematopoietic grafts could lead to improved strategies for the development of cellular therapies, for example in situations where platelet engraftment is delayed.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Adolescente , Adulto , Fatores Etários , Idoso , Antígenos CD34/imunologia , Doadores de Sangue , Transplante de Medula Óssea , Contagem de Células , Células Precursoras Eritroides/imunologia , Sangue Fetal/imunologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Humanos , Pessoa de Meia-Idade , Agonistas Mieloablativos/uso terapêutico
16.
Exp Hematol ; 39(1): 26-36.e1, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20974210

RESUMO

OBJECTIVE: These studies were performed to test the hypothesis that DNMT1 is required for maintenance of DNA methylation and repression of the γ-globin gene in adult-stage erythroid cells. MATERIALS AND METHODS: DNMT1 levels were reduced by nucleofection of small interfering RNA targeting DNMT1 in chemical inducer of dimerization-dependent multipotential mouse bone marrow cells containing the human ß-globin gene locus in the context of a yeast artificial chromosome and in primary cultures of erythroid progenitor cells derived from CD34(+) baboon bone marrow cells. The effect of reduced DNMT1 levels on globin gene expression was measured by real-time polymerase chain reaction and the effect on globin chain synthesis in primary erythroid progenitor cell cultures was determined by biosynthetic radiolabeling of globin chains followed by high-performance liquid chromatography analysis. The effect on DNA methylation was determined by bisulfite sequence analysis. RESULTS: Reduced DNMT1 levels in cells treated with siDNMT1 were associated with increased expression of γ-globin messenger RNA, an increased γ/γ+ß chain ratio in cultured erythroid progenitors, and decreased DNA methylation of the γ-globin promoter. Similar effects were observed in cells treated with decitabine, a pharmacological inhibitor of DNA methyltransferase inhibitor. CONCLUSIONS: DNMT1 is required to maintain DNA methylation of the γ-globin gene promoter and repress γ-globin gene expression in adult-stage erythroid cells.


Assuntos
Células da Medula Óssea/metabolismo , DNA (Citosina-5-)-Metiltransferases/fisiologia , Células Precursoras Eritroides/metabolismo , gama-Globinas/genética , Animais , Antígenos CD34/imunologia , Western Blotting , Células Cultivadas , Cromossomos Artificiais de Levedura , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Dimerização , Células Precursoras Eritroides/imunologia , Humanos , Camundongos , Papio , RNA Interferente Pequeno/genética , Transfecção
17.
J Immunol ; 186(1): 203-13, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21115735

RESUMO

Signal transduction mediated by Fas-associated death domain protein (FADD) represents a paradigm of coregulation of apoptosis and cellular proliferation. During apoptotic signaling induced by death receptors including Fas, FADD is required for the recruitment and activation of caspase 8. In addition, a death receptor-independent function of FADD is essential for embryogenesis. In previous studies, FADD deficiency in embryonic stem cells resulted in a complete lack of B cells and dramatically reduced T cell numbers, as shown by Rag1(-/-) blastocyst complementation assays. However, T-specific FADD-deficient mice contained normal numbers of thymocytes and slightly reduced peripheral T cell numbers, whereas B cell-specific deletion of FADD led to increased peripheral B cell numbers. It remains undetermined what impact an FADD deficiency has on hematopoietic stem cells and progenitors. The current study analyzed the effect of simultaneous deletion of FADD in multiple cell types, including bone marrow cells, by using the IFN-inducible Mx1-cre transgene. The resulting FADD mutant mice did not develop lymphoproliferation diseases, unlike Fas-deficient mice. Instead, a time-dependent depletion of peripheral FADD-deficient lymphocytes was observed. In the bone marrow, a lack of FADD led to a dramatic decrease in the hematopoietic stem cells and progenitor-enriched population. Furthermore, FADD-deficient bone marrow cells were defective in their ability to generate lymphoid, myeloid, and erythroid cells. Thus, the results revealed a temporal requirement for FADD. Although dispensable during lymphopoiesis post lineage commitment, FADD plays a critical role in early hematopoietic stages in the bone marrow.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Proteína de Domínio de Morte Associada a Fas/deficiência , Proteína de Domínio de Morte Associada a Fas/genética , Hematopoese/genética , Hematopoese/imunologia , Depleção Linfocítica/métodos , Animais , Células da Medula Óssea/metabolismo , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Células Cultivadas , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Proteínas de Ligação ao GTP/genética , Deleção de Genes , Proteínas de Fluorescência Verde/genética , Integrases/genética , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Proteínas de Resistência a Myxovirus , Fatores de Tempo , Receptor fas/deficiência , Receptor fas/genética
18.
Pediatr Dev Pathol ; 13(4): 273-81, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19863449

RESUMO

The presence of intrauterine inflammation has been associated with adverse neurologic outcomes in preterm infants, but the precise mechanisms of fetal brain injury remain unclear. We sought to evaluate inflammatory cell trafficking, fetal organ damage, and molecular regulation in the fetoplacental unit using an established mouse model of preterm birth associated with intrauterine inflammation. Gestational sacs were harvested 6 hours after intrauterine infusion of saline or lipopolysaccharide (LPS). Histologic, immunohistochemical, and molecular investigations were performed to identify target organ damage and the cellular phenotype of inflammatory cells and to quantify circulating inflammatory and hematopoietic mediators within the placental and fetal tissue. There was widespread increase in fetal macrophages in LPS-exposed pups, including within the leptomeninges of the brain, associated with significantly higher of interleukin 6 levels in LPS-exposed pups. Although no specific central nervous system injury (necrosis or apoptosis) was documented, liver hematomas were seen significantly more frequently in LPS-exposed pups. Circulating nucleated fetal erythrocytes were also present more frequently with LPS exposure without significantly higher erythropoietin levels than saline-exposed mice. The presence of increased macrophages, increased circulating interleukin 6 levels, and increased circulating erythroid precursors in LPS-exposed pups suggests that these are significant factors associated with potential target organ damage, such as liver hematomas, associated with intrauterine inflammation and preterm birth. The role of macrophages within the fetal leptomeninges is unclear, but they may play an important role in inflammatory-mediated brain damage, and further investigation of their significance and potential as therapeutic targets is warranted.


Assuntos
Encefalopatias/embriologia , Modelos Animais de Doenças , Células Precursoras Eritroides/imunologia , Doenças Fetais/imunologia , Macrófagos/imunologia , Complicações Infecciosas na Gravidez , Nascimento Prematuro/imunologia , Animais , Animais não Endogâmicos , Biomarcadores/metabolismo , Feminino , Inflamação/imunologia , Interleucina-6/sangue , Lipopolissacarídeos , Camundongos , Gravidez
19.
Clin Chem Lab Med ; 48(2): 199-203, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20001441

RESUMO

BACKGROUND: Exercise in humans augments the mobilization of circulating hematopoietic progenitor cells (CD34(+)) from the bone marrow. We investigated the effect of inflammation on erythroid marrow activity by mobilization of erythroid progenitor cells (EPs) along with soluble markers of erythropoiesis. METHODS: Ten healthy athletes who participated in an ultradistance foot race participated in the study. Peripheral blood mononuclear cells were isolated, before (phase I), at the end (phase II), and at 48 h post-race (phase III). EPs were detected as burst colony forming units (BFU-e) and colonies were scored at day 14. Markers of inflammation (C-reactive protein, serum amyloid-A, interleukin-6, ferritin and S100B) and bone marrow activity (erythropoietin, soluble transferrin receptor and lipocalin-2) were assessed. RESULTS: An approximately three-fold decrease in BFU-e number was observed at phase II. sTfR concentrations were also decreased at phase II and remained decreased at phase III. However, EPO and lipocalin-2 concentrations reached a maximum value at phase II, with a tendency to decrease at phase III. CONCLUSIONS: These findings indicate that exercise-induced inflammation modulates bone marrow homeostasis leading to an increase in leukocyte turnover and a decrease in erythroid compartment. It appears that lipocalin-2 is the main factor that regulates the production and mobilization of EPs.


Assuntos
Biomarcadores/sangue , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Eritropoese/fisiologia , Inflamação/sangue , Esforço Físico/fisiologia , Corrida/lesões , Proteínas de Fase Aguda/imunologia , Proteínas de Fase Aguda/metabolismo , Atletas , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Células Precursoras Eritroides/imunologia , Eritropoese/imunologia , Eritropoetina/sangue , Ferritinas/sangue , Humanos , Inflamação/etiologia , Inflamação/imunologia , Lipocalina-2 , Lipocalinas/imunologia , Lipocalinas/metabolismo , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores da Transferrina/sangue , Fatores de Tempo
20.
Pediatr Hematol Oncol ; 26(5): 338-44, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19579080

RESUMO

Thalassemic children are at a high risk of graft rejection in cord blood transplantation. To investigate this possible mechanism, the authors evaluated the effect of panel reactive antibody on the growth of CD34(+) cells in vitro. On semisolid medium, CD34(+) cells derived from cord blood were incubated with thalassemic serum, and colony-forming units were counted after 10 days of culture. After incubation with serum-specific panel reactive antibody, profound decreases were found in the numbers of CFU-GM, CFU-GEMM and BFU-E compared with controls. The results indicated that serum-specific panel reactive antibody might have an apparent inhibition effect on proliferation and differentiation of cord blood CD34(+) cells.


Assuntos
Anticorpos/imunologia , Antígenos CD34/imunologia , Células Precursoras Eritroides/imunologia , Sangue Fetal/imunologia , Células Progenitoras de Granulócitos e Macrófagos/imunologia , Células Progenitoras Mieloides/imunologia , Talassemia beta/imunologia , Anticorpos/sangue , Apoptose/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Criança , Células Precursoras Eritroides/citologia , Sangue Fetal/citologia , Células Progenitoras de Granulócitos e Macrófagos/citologia , Humanos , Células Progenitoras Mieloides/citologia , Talassemia beta/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA