Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 49(4): 627-639.e6, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30314756

RESUMO

The non-hematopoietic cell fraction of the bone marrow (BM) is classically identified as CD45- Ter119- CD31- (herein referred to as triple-negative cells or TNCs). Although TNCs are believed to contain heterogeneous stromal cell populations, they remain poorly defined. Here we showed that the vast majority of TNCs (∼85%) have a hematopoietic rather than mesenchymal origin. Single cell RNA-sequencing revealed erythroid and lymphoid progenitor signatures among CD51- TNCs. Ly6D+ CD44+ CD51- TNCs phenotypically and functionally resembled CD45+ pro-B lymphoid cells, whereas Ly6D- CD44+ CD51- TNCs were enriched in previously unappreciated stromal-dependent erythroid progenitors hierarchically situated between preCFU-E and proerythroblasts. Upon adoptive transfer, CD44+ CD51- TNCs contributed to repopulate the B-lymphoid and erythroid compartments. CD44+ CD51- TNCs also expanded during phenylhydrazine-induced acute hemolysis or in a model of sickle cell anemia. These findings thus uncover physiologically relevant new classes of stromal-associated functional CD45- hematopoietic progenitors.


Assuntos
Células da Medula Óssea/imunologia , Células Eritroides/imunologia , Células Progenitoras Linfoides/imunologia , Células Estromais/imunologia , Transferência Adotiva/métodos , Animais , Antígenos de Grupos Sanguíneos/imunologia , Antígenos de Grupos Sanguíneos/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Células Eritroides/citologia , Células Eritroides/metabolismo , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo
2.
Arch Virol ; 169(5): 103, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38632180

RESUMO

Missense mutations in certain small envelope proteins diminish the efficacy of antibodies. Consequently, tracking the incidence and types of vaccine-escape mutations (VEMs) was crucial both before and after the introduction of universal hepatitis B vaccination in Japan in 2016. In this study, we isolated hepatitis B virus (HBV) DNA from 58 of 169 hepatitis B surface antigen (HBsAg)-positive blood samples from Japanese blood donors and determined the nucleotide sequence encoding the small envelope protein. DNA from six (10%) of the samples had VEMs, but no missense mutations, such as G145R, were detected. Complete HBV genome sequences were obtained from 29 of the 58 samples; the viral genotype was A1 in one (3%), A2 in three (10%), B1 in nine (31%), B2 in five (17%), B4 in one (3%), and C2 in 10 (34%) samples. Tenofovir-resistance mutations were detected in two (7%) samples. In addition, several core promoter mutations, such as 1762A>T and 1764G>A, and a precore nonsense mutation, 1986G>A, which are risk factors for HBV-related chronic liver disease, were detected. These findings provide a baseline for future research and highlight the importance of ongoing monitoring of VEMs and drug resistance mutations in HBV DNA from HBsAg-positive blood donors without HBV antibodies.


Assuntos
Hepatite B Crônica , Hepatite B , Humanos , Vírus da Hepatite B/genética , Antígenos de Superfície da Hepatite B/genética , Japão , Doadores de Sangue , DNA Viral/genética , Mutação , Genótipo
3.
Blood ; 138(24): 2555-2569, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34587247

RESUMO

Neutrophils play an essential role in innate immune responses to bacterial and fungal infections, and loss of neutrophil function can increase the risk of acquiring lethal infections in clinical settings. Here, we show that engineered neutrophil-primed progenitors derived from human induced pluripotent stem cells can produce functional neutrophil-like cells at a clinically applicable scale that can act rapidly in vivo against lethal bacterial infections. Using 5 different mouse models, we systematically demonstrated that these neutrophil-like cells migrate to sites of inflammation and infection and increase survival against bacterial infection. In addition, we found that these human neutrophil-like cells can recruit murine immune cells. This system potentially provides a straight-forward solution for patients with neutrophil deficiency: an off-the-shelf neutrophil transfusion. This platform should facilitate the administration of human neutrophils for a broad spectrum of physiological and pathological conditions.


Assuntos
Infecções Bacterianas/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Neutrófilos/transplante , Animais , Infecções Bacterianas/imunologia , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imunidade Inata , Células-Tronco Pluripotentes Induzidas/imunologia , Inflamação/imunologia , Inflamação/terapia , Camundongos Endogâmicos BALB C , Neutrófilos/citologia , Neutrófilos/imunologia
4.
Immunity ; 37(5): 827-39, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23123064

RESUMO

Mast cells (MCs) are key effector cells in allergic reactions. However, the inhibitory mechanism that prevents excessive activation of MCs remains elusive. Here we show that leukocyte mono-immunoglobulin-like receptor 3 (LMIR3; also called CD300f) is a negative regulator of MC activation in vivo. LMIR3 deficiency exacerbated MC-dependent allergic responses in mice, including anaphylaxis, airway inflammation, and dermatitis. Both physical binding and functional reporter assays via an extracellular domain of LMIR3 showed that several extracellular lipids (including ceramide) and lipoproteins were possible ligands for LMIR3. Importantly, MCs were frequently surrounded by extracellular ceramide in vivo. Upon engagement of high-affinity immunoglobulin E receptor, extracellular ceramide-LMIR3 binding inhibited MC activation via immunoreceptor tyrosine-based inhibitory and switch motifs of LMIR3. Moreover, pretreatment with LMIR3-Fc fusion protein or antibody against either ceramide or LMIR3 interfered with this binding in vivo, thereby exacerbating passive cutaneous anaphylaxis. Thus, the interaction between extracellular ceramide and LMIR3 suppressed MC-dependent allergic responses.


Assuntos
Ceramidas/imunologia , Ceramidas/metabolismo , Hipersensibilidade/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Anafilaxia/imunologia , Anafilaxia/metabolismo , Animais , Células Cultivadas , Dermatite/imunologia , Dermatite/metabolismo , Hipersensibilidade/metabolismo , Hipersensibilidade/patologia , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Lipoproteínas/imunologia , Lipoproteínas/metabolismo , Mastócitos/patologia , Camundongos , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Receptores de IgE/imunologia , Receptores de IgE/metabolismo , Tirosina/imunologia , Tirosina/metabolismo
6.
Blood ; 123(25): 3932-42, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24825862

RESUMO

High levels of HES1 expression are frequently found in BCR-ABL(+) chronic myelogenous leukemia in blast crisis (CML-BC). In mouse bone marrow transplantation (BMT) models, co-expression of BCR-ABL and Hes1 induces CML-BC-like disease; however, the underlying mechanism remained elusive. Here, based on gene expression analysis, we show that MMP-9 is upregulated by Hes1 in common myeloid progenitors (CMPs). Analysis of promoter activity demonstrated that Hes1 upregulated MMP-9 by activating NF-κB. Analysis of 20 samples from CML-BC patients showed that MMP-9 was highly expressed in three, with two exhibiting high levels of HES1 expression. Interestingly, MMP-9 deficiency impaired the cobblestone area-forming ability of CMPs expressing BCR-ABL and Hes1 that were in conjunction with a stromal cell layer. In addition, CMPs expressing BCR-ABL and Hes1 secreted MMP-9, promoting the release of soluble Kit-ligand (sKitL) from stromal cells, thereby enhancing proliferation of the leukemic cells. In accordance, mice transplanted with CMPs expressing BCR-ABL and Hes1 exhibited high levels of sKitL as well as MMP-9 in the serum. Importantly, MMP-9 deficiency impaired the development of CML-BC-like disease induced by BCR-ABL and Hes1 in mouse BMT models. The present results suggest that Hes1 promotes the development of CML-BC, partly through MMP-9 upregulation in leukemic cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Crise Blástica/genética , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Metaloproteinase 9 da Matriz/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Crise Blástica/metabolismo , Transplante de Medula Óssea/métodos , Movimento Celular/genética , Proliferação de Células , Citometria de Fluxo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Estimativa de Kaplan-Meier , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição HES-1 , Regulação para Cima
7.
J Biol Chem ; 288(11): 7662-7675, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23372157

RESUMO

CD300C is highly homologous with an inhibitory receptor CD300A in an immunoglobulin-like domain among the human CD300 family of paired immune receptors. To clarify the precise expression and function of CD300C, we generated antibodies discriminating between CD300A and CD300C, which recognized a unique epitope involving amino acid residues CD300A(F56-L57) and CD300C(L63-R64). Notably, CD300C was highly expressed in human monocytes and mast cells. Cross-linking of CD300C by its specific antibody caused cytokine/chemokine production of human monocytes and mast cells. Fc receptor γ was indispensable for both efficient surface expression and activating functions of CD300C. To identify a ligand for CD300A or CD300C, we used reporter cell lines expressing a chimera receptor harboring extracellular CD300A or CD300C and intracellular CD3ζ, in which its unknown ligand induced GFP expression. Our results indicated that phosphatidylethanolamine (PE) among the lipids tested and apoptotic cells were possible ligands for both CD300C and CD300A. PE and apoptotic cells more strongly induced GFP expression in the reporter cells through binding to extracellular CD300A as compared with CD300C. Differential recognition of PE by extracellular CD300A and CD300C depended on different amino acid residues CD300A(F56-L57) and CD300C(L63-R64). Interestingly, GFP expression induced by extracellular CD300C-PE binding in the reporter cells was dampened by co-expression of full-length CD300A, indicating the predominance of CD300A over CD300C in PE recognition/signaling. PE consistently failed to stimulate cytokine production in monocytes expressing CD300C with CD300A. In conclusion, specific engagement of CD300C led to Fc receptor γ-dependent activation of mast cells and monocytes.


Assuntos
Antígenos de Superfície/fisiologia , Regulação da Expressão Gênica , Mastócitos/metabolismo , Glicoproteínas de Membrana/fisiologia , Monócitos/metabolismo , Receptores de IgG/metabolismo , Animais , Antígenos de Superfície/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Células HL-60 , Humanos , Sistema Imunitário , Células Jurkat , Células K562 , Ligantes , Mastócitos/citologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Células NIH 3T3 , Fagocitose , Ratos , Transdução de Sinais , Relação Estrutura-Atividade , Células U937
8.
J Immunol ; 189(4): 1773-9, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22772446

RESUMO

Leukocyte mono-Ig-like receptor 5 (LMIR5, also called CD300b) is an activating receptor expressed in myeloid cells. We have previously demonstrated that T cell Ig mucin 1 works as a ligand for LMIR5 in mouse ischemia/reperfusion injury of the kidneys. In this article, we show that LMIR5 is implicated in LPS-induced sepsis in mice. Notably, neutrophils constitutively released a soluble form of LMIR5 (sLMIR5) through proteolytic cleavage of surface LMIR5. Stimulation with TLR agonists augmented the release of sLMIR5. LPS administration or peritonitis induction increased serum levels of sLMIR5 in mice, which was substantially inhibited by neutrophil depletion. Thus, neutrophils were the main source of LPS-induced sLMIR5 in vivo. On the other hand, i.p. administration of LMIR5-Fc, a surrogate of sLMIR5, bound to resident macrophages (M) and stimulated transient inflammation in mice. Consistently, LMIR5-Fc induced in vitro cytokine production of peritoneal M via its unknown ligand. Interestingly, LMIR5 deficiency profoundly reduced systemic cytokine production and septic mortality in LPS-administered mice, although it did not affect in vitro cytokine production of LPS-stimulated peritoneal M. Importantly, the resistance of LMIR5-deficient mice to LPS- or peritonitis-induced septic death was decreased by LMIR5-Fc administration, implicating sLMIR5 in LPS responses in vivo. Collectively, neutrophil-derived sLMIR5 amplifies LPS-induced lethal inflammation.


Assuntos
Neutrófilos/imunologia , Receptores Imunológicos/imunologia , Sepse/imunologia , Animais , Western Blotting , Citocinas/biossíntese , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Inflamação/induzido quimicamente , Inflamação/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos/genética , Sepse/induzido quimicamente , Solubilidade , Transfecção
9.
Proc Jpn Acad Ser B Phys Biol Sci ; 90(10): 389-404, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25504228

RESUMO

Myeloid malignancies consist of acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and myeloproliferative neoplasm (MPN). The latter two diseases have preleukemic features and frequently evolve to AML. As with solid tumors, multiple mutations are required for leukemogenesis. A decade ago, these gene alterations were subdivided into two categories: class I mutations stimulating cell growth or inhibiting apoptosis; and class II mutations that hamper differentiation of hematopoietic cells. In mouse models, class I mutations such as the Bcr-Abl fusion kinase induce MPN by themselves and some class II mutations such as Runx1 mutations induce MDS. Combinations of class I and class II mutations induce AML in a variety of mouse models. Thus, it was postulated that hematopoietic cells whose differentiation is blocked by class II mutations would autonomously proliferate with class I mutations leading to the development of leukemia. Recent progress in high-speed sequencing has enabled efficient identification of novel mutations in a variety of molecules including epigenetic factors, splicing factors, signaling molecules and proteins in the cohesin complex; most of these are not categorized as either class I or class II mutations. The functional consequences of these mutations are now being extensively investigated. In this article, we will review the molecular basis of hematological malignancies, focusing on mouse models and the interfaces between these models and clinical findings, and revisit the classical class I/II hypothesis.


Assuntos
Transformação Celular Neoplásica/genética , Epigênese Genética , Neoplasias Hematológicas/genética , Leucemia Mieloide Aguda/genética , Mutação , Síndromes Mielodisplásicas/genética , Animais , Proliferação de Células/genética , Transformação Celular Neoplásica/metabolismo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Neoplasias Hematológicas/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Camundongos , Síndromes Mielodisplásicas/metabolismo
10.
Exp Hematol ; 130: 104137, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38103826

RESUMO

Neutrophils are key components of the immune system that inhibit bacterial infections. Systemic bacterial infections can cause lethal conditions, especially in patients with neutropenia associated with chemotherapy or other systemic illnesses; hence, early detection of the symptoms and prompt management are crucial in such cases. Previously, we established expandable engineered neutrophil-primed progenitors (NeuPs-XL) using human-induced pluripotent stem cells (iPSCs), which can produce neutrophil-like cells at a clinically suitable scale within 4 days of inducing myeloid differentiation. In this study, using small-molecule compound-based screening, we detected that MK-2206, a selective pan-AKT inhibitor, can accelerate this differentiation process, promote phagocytic ability in neutrophils, and enhance cytokine and chemokine expression in response to lipopolysaccharides. The inhibition of AKT2 has been identified as the key mechanism underlying this acceleration. These results can make a substantial contribution to the development of strategies for the prompt production of clinically applicable iPSC-derived neutrophils, which can potentially lead to the management of severe infections associated with life-threatening neutropenia and the effective treatment of related health conditions in the future.


Assuntos
Infecções Bacterianas , Células-Tronco Pluripotentes Induzidas , Neutropenia , Humanos , Neutrófilos/metabolismo , Diferenciação Celular , Neutropenia/metabolismo , Infecções Bacterianas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
Blood ; 117(1): 221-33, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20884804

RESUMO

Two types of mutations of a transcription factor CCAAT-enhancer binding protein α (C/EBPα) are found in leukemic cells of 5%-14% of acute myeloid leukemia (AML) patients: N-terminal mutations expressing dominant negative p30 and C-terminal mutations in the basic leucine zipper domain. Our results showed that a mutation of C/EBPα in one allele was observed in AML after myelodysplastic syndrome, while the 2 alleles are mutated in de novo AML. Unlike an N-terminal frame-shift mutant (C/EBPα-N(m))-transduced cells, a C-terminal mutant (C/EBPα-C(m))-transduced cells alone induced AML with leukopenia in mice 4-12 months after bone marrow transplantation. Coexpression of both mutants induced AML with marked leukocytosis with shorter latencies. Interestingly, C/EBPα-C(m) collaborated with an Flt3-activating mutant Flt3-ITD in inducing AML. Moreover, C/EBPα-C(m) strongly blocked myeloid differentiation of 32Dcl3 cells, suggesting its class II mutation-like role in leukemogenesis. Although C/EBPα-C(m) failed to inhibit transcriptional activity of wild-type C/EBPα, it suppressed the synergistic effect between C/EBPα and PU.1. On the other hand, C/EBPα-N(m) inhibited C/EBPα activation in the absence of PU.1, despite low expression levels of p30 protein generated by C/EBPα-N(m). Thus, 2 types of C/EBPα mutations are implicated in leukemo-genesis, involving different and cooperating molecular mechanisms.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/genética , Modelos Animais de Doenças , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/patologia , Mutação/genética , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Western Blotting , Transplante de Medula Óssea , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Diferenciação Celular , Proliferação de Células , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Citometria de Fluxo , Humanos , Leucemia Mieloide Aguda/terapia , Luciferases/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/terapia , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , RNA Mensageiro/genética , Taxa de Sobrevida , Ativação Transcricional
12.
Cancer Sci ; 103(1): 26-33, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21943131

RESUMO

Transforming growth factor-ß (TGF-ß)-stimulated clone-22 (TSC-22), also called TSC22D1-2, is a putative tumor suppressor. We previously identified TSC-22 downstream of an active mutant of fms-like tyrosine kinase-3 (Flt3). Here, we show that TSC-22 works as a tumor suppressor through inhibiting Ras/Raf signaling. Notably, TSC-22 was upregulated by Ras/Raf activation, whereas its upregulation was inhibited by concurrent STAT5 activation. Although TSC-22 was normally retained in the cytoplasm by its nuclear export signal (NES), Ras/Raf activation caused nuclear translocation of TSC-22, but not TSC22D1-1. Unlike glucocorticoid-induced leucine zipper (GILZ/TSC22D3-2) previously characterized as a negative regulator of Ras/Raf signaling, TSC-22 failed to interact physically with Ras/Raf. Importantly, transduction with TSC-22, but not TSC22D1-1, suppressed the growth, transformation and tumorigenesis of NIH3T3 cells expressing oncogenic H-Ras: this suppression was enhanced by transduction with a TSC-22 mutant lacking NES that had accumulated in the nucleus. Collectively, upregulation and nuclear translocation of TSC-22 played an important role in the feedback suppression of Ras/Raf signaling. Consistently, TSC22D1-deficient mice were susceptible to tumorigenesis in a mouse model of chemically-induced liver tumors bearing active mutations of Ras/Raf. Thus, TSC-22 negatively regulated Ras/Raf signaling through a mechanism different from GILZ, implicating TSC-22 as a novel suppressor of oncogenic Ras/Raf-induced tumors.


Assuntos
Transformação Celular Neoplásica/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas Experimentais/patologia , Proteínas Repressoras/fisiologia , Quinases raf/metabolismo , Proteínas ras/metabolismo , Animais , Western Blotting , Células Cultivadas , Dietilnitrosamina/toxicidade , Imunoprecipitação , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Células Precursoras de Linfócitos B , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Quinases raf/genética , Proteínas ras/genética
13.
Blood ; 115(14): 2872-81, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19861684

RESUMO

Hairy enhancer of split 1 (Hes1) is a basic helix-loop-helix transcriptional repressor that affects differentiation and often helps maintain cells in an immature state in various tissues. Here we show that retroviral expression of Hes1 immortalizes common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) in the presence of interleukin-3, conferring permanent replating capability on these cells. Whereas these cells did not develop myeloproliferative neoplasms when intravenously administered to irradiated mice, the combination of Hes1 and BCR-ABL in CMPs and GMPs caused acute leukemia resembling blast crisis of chronic myelogenous leukemia (CML), resulting in rapid death of the recipient mice. On the other hand, BCR-ABL alone caused CML-like disease when expressed in c-Kit-positive, Sca-1-positive, and lineage-negative hematopoietic stem cells (KSLs), but not committed progenitors CMPs or GMPs, as previously reported. Leukemic cells derived from Hes1 and BCR-ABL-expressing CMPs and GMPs were more immature than those derived from BCR-ABL-expressing KSLs. Intriguingly, Hes1 was highly expressed in 8 of 20 patients with CML in blast crisis, but not in the chronic phase, and dominant negative Hes1 retarded the growth of some CML cell lines expressing Hes1. These results suggest that Hes1 is a key molecule in blast crisis transition in CML.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Crise Blástica/metabolismo , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Proteínas de Homeodomínio/biossíntese , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Repressoras/biossíntese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Crise Blástica/genética , Crise Blástica/patologia , Linhagem Celular Transformada , Proteínas de Fusão bcr-abl/biossíntese , Proteínas de Fusão bcr-abl/genética , Células Progenitoras de Granulócitos e Macrófagos/patologia , Proteínas de Homeodomínio/genética , Interleucina-3/genética , Interleucina-3/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Células-Tronco Neoplásicas/patologia , Ratos , Proteínas Repressoras/genética , Fatores de Transcrição HES-1
14.
Nat Cell Biol ; 24(3): 290-298, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35210567

RESUMO

Haematopoietic stem cells (HSCs) home to the bone marrow via, in part, interactions with vascular cell adhesion molecule-1 (VCAM1)1-3. Once in the bone marrow, HSCs are vetted by perivascular phagocytes to ensure their self-integrity. Here we show that VCAM1 is also expressed on healthy HSCs and upregulated on leukaemic stem cells (LSCs), where it serves as a quality-control checkpoint for entry into bone marrow by providing 'don't-eat-me' stamping in the context of major histocompatibility complex class-I (MHC-I) presentation. Although haplotype-mismatched HSCs can engraft, Vcam1 deletion, in the setting of haplotype mismatch, leads to impaired haematopoietic recovery due to HSC clearance by mononuclear phagocytes. Mechanistically, VCAM1 'don't-eat-me' activity is regulated by ß2-microglobulin MHC presentation on HSCs and paired Ig-like receptor-B (PIR-B) on phagocytes. VCAM1 is also used by cancer cells to escape immune detection as its expression is upregulated in multiple cancers, including acute myeloid leukaemia (AML), where high expression associates with poor prognosis. In AML, VCAM1 promotes disease progression, whereas VCAM1 inhibition or deletion reduces leukaemia burden and extends survival. These results suggest that VCAM1 engagement regulates a critical immune-checkpoint gate in the bone marrow, and offers an alternative strategy to eliminate cancer cells via modulation of the innate immune tolerance.


Assuntos
Leucemia Mieloide Aguda , Molécula 1 de Adesão de Célula Vascular , Medula Óssea , Células-Tronco Hematopoéticas/metabolismo , Humanos , Tolerância Imunológica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Neoplásicas , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
15.
J Immunol ; 182(10): 6168-78, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414770

RESUMO

The development of NK cells from hematopoietic stem cells is thought to be dependent on IL-15. In this study, we demonstrate that stimulation of human cord blood CD34(+) cells by a Notch ligand, Delta4, along with IL-7, stem cell factor, and Fms-like tyrosine kinase 3 ligand, but no IL-15, in a stroma-free culture induced the generation of cells with characteristics of functional NK cells, including CD56 and CD161 Ag expression, IFN-gamma secretion, and cytotoxic activity against K562 and Jurkat cells. Addition of gamma-secretase inhibitor and anti-human Notch1 Ab to the culture medium almost completely blocked NK cell emergence. Addition of anti-human IL-15-neutralizing Ab did not affect NK cell development in these culture conditions. The presence of IL-15, however, augmented cytotoxicity and was required for a more mature NK cell phenotype. CD56(+) cells generated by culture with IL-15, but without Notch stimulation, were negative for CD7 and cytoplasmic CD3, whereas CD56(+) cells generated by culture with both Delta4 and IL-15 were CD7(+) and cytoplasmic CD3(+) from the beginning and therefore more similar to in vivo human NK cell progenitors. Together, these results suggest that Notch signaling is important for the physiologic development of NK cells at differentiation stages beyond those previously postulated.


Assuntos
Diferenciação Celular/imunologia , Células-Tronco Hematopoéticas/citologia , Interleucina-15/metabolismo , Células Matadoras Naturais/citologia , Subpopulações de Linfócitos/citologia , Receptores Notch/metabolismo , Antígenos CD34/imunologia , Antígenos CD34/metabolismo , Sangue Fetal , Citometria de Fluxo , Imunofluorescência , Células-Tronco Hematopoéticas/imunologia , Humanos , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Receptores Notch/imunologia , Transdução de Sinais/imunologia
16.
Stem Cell Reports ; 16(12): 2871-2886, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34798065

RESUMO

Hematopoietic cells differentiate through several progenitors in a hierarchical manner, and recent single-cell analyses have revealed substantial heterogeneity within each progenitor. Although common myeloid progenitors (CMPs) are defined as a multipotent cell population that can differentiate into granulocyte-monocyte progenitors (GMPs) and megakaryocyte-erythrocyte progenitors (MEPs), and GMPs generate neutrophils and monocytes, these myeloid progenitors must contain some lineage-committed progenitors. Through gene expression analysis at single-cell levels, we identified CD62L as a marker to reveal the heterogeneity. We confirmed that CD62L-negative CMPs represent "bona fide" CMPs, whereas CD62L-high CMPs are mostly restricted to GMP potentials both in mice and humans. In addition, we identified CD62L-negative GMPs as the most immature subsets in GMPs and Ly6C+CD62L-intermediate and Ly6C+CD62L-high GMPs are skewed to neutrophil and monocyte differentiation in mice, respectively. Our findings contribute to more profound understanding about the mechanism of myeloid differentiation.


Assuntos
Linhagem da Célula , Selectina L/metabolismo , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo , Animais , Diferenciação Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo
17.
Leuk Lymphoma ; 62(3): 614-619, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33076723

RESUMO

Although anti-interleukin-6 therapy with tocilizumab and siltuximab is recommended for multicentric Castleman's disease (MCD), burdens caused by frequent hospital visits and high drug payments is an issue to be considered. Although glucocorticoid monotherapy might be less effective compared to these agents, substantial proportions of patients can be successfully treated for years. Therefore, we conducted a retrospective analysis of Castleman's disease patients to explore predictors of glucocorticoid responsiveness and revealed that higher hemoglobin and/or lower C-reactive protein levels before starting glucocorticoid monotherapy were associated with lower probability of requirements for second-line treatment among patients initially treated with glucocorticoid. We concluded that glucocorticoids had a potential to induce sustained disease control for indolent MCD patients with specific clinical characteristics.


Assuntos
Hiperplasia do Linfonodo Gigante , Proteína C-Reativa , Hiperplasia do Linfonodo Gigante/diagnóstico , Hiperplasia do Linfonodo Gigante/tratamento farmacológico , Glucocorticoides/uso terapêutico , Hemoglobinas , Humanos , Estudos Retrospectivos
18.
Intern Med ; 60(18): 2953-2959, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-33814498

RESUMO

The risk of carcinogenesis increases after 20 years old in patients with Fanconi anemia (FA). We herein report three rare cases of FA combined with esophageal cancer in women; all patients were diagnosed with FA in early childhood. Patients 1 and 2 were diagnosed with advanced and superficial esophageal cancer, respectively, at 21 and 30 years old, respectively. Patient 3 was diagnosed with superficial esophageal cancer, underwent curative surgery at 26 years old, and survived for over 5 years without recurrence. Therefore, establishing a protocol for the early detection of esophageal cancer in FA patients over 20 years old is important.


Assuntos
Neoplasias Esofágicas , Anemia de Fanconi , Adulto , Neoplasias Esofágicas/complicações , Neoplasias Esofágicas/diagnóstico , Anemia de Fanconi/complicações , Anemia de Fanconi/diagnóstico , Feminino , Humanos , Recidiva Local de Neoplasia , Adulto Jovem
19.
Intern Med ; 60(2): 217-222, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-32921688

RESUMO

At 37 years old, a patient developed chronic watery diarrhea, generalized pain, severe hypokalemia and elevated creatine kinase levels. She was thought to have rhabdomyolysis due to hypokalemia from chronic diarrhea. No organic cause was found. Her symptoms subsided with potassium correction, but hypokalemia persisted; she visited our hospital at 44 years old. Endoscopy detected prominent atrophy of the intestinal villi. Histology indicated Marsh-Oberhuber type-3b disease. Anti-gliadin and anti-tissue transglutaminase IgA antibody tests were positive. She was diagnosed with celiac disease and started on a gluten-free diet, which improved her symptoms. This report is only the tenth of its kind worldwide.


Assuntos
Doença Celíaca , Rabdomiólise , Adulto , Doença Celíaca/complicações , Doença Celíaca/diagnóstico , Duodeno , Feminino , Gliadina , Humanos , Imunoglobulina A , Mucosa Intestinal , Rabdomiólise/complicações , Rabdomiólise/diagnóstico
20.
Nat Commun ; 12(1): 2522, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33947846

RESUMO

Haematopoietic stem cells (HSCs) tightly regulate their quiescence, proliferation, and differentiation to generate blood cells during the entire lifetime. The mechanisms by which these critical activities are balanced are still unclear. Here, we report that Macrophage-Erythroblast Attacher (MAEA, also known as EMP), a receptor thus far only identified in erythroblastic island, is a membrane-associated E3 ubiquitin ligase subunit essential for HSC maintenance and lymphoid potential. Maea is highly expressed in HSCs and its deletion in mice severely impairs HSC quiescence and leads to a lethal myeloproliferative syndrome. Mechanistically, we have found that the surface expression of several haematopoietic cytokine receptors (e.g. MPL, FLT3) is stabilised in the absence of Maea, thereby prolonging their intracellular signalling. This is associated with impaired autophagy flux in HSCs but not in mature haematopoietic cells. Administration of receptor kinase inhibitor or autophagy-inducing compounds rescues the functional defects of Maea-deficient HSCs. Our results suggest that MAEA provides E3 ubiquitin ligase activity, guarding HSC function by restricting cytokine receptor signalling via autophagy.


Assuntos
Autofagossomos/genética , Autofagia/genética , Moléculas de Adesão Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Proteínas do Citoesqueleto/genética , Perfilação da Expressão Gênica , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Estabilidade Proteica , Receptores de Trombopoetina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Tirosina Quinase 3 Semelhante a fms/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA