Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 172(1-2): 147-161.e12, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29328910

RESUMO

Trained innate immunity fosters a sustained favorable response of myeloid cells to a secondary challenge, despite their short lifespan in circulation. We thus hypothesized that trained immunity acts via modulation of hematopoietic stem and progenitor cells (HSPCs). Administration of ß-glucan (prototypical trained-immunity-inducing agonist) to mice induced expansion of progenitors of the myeloid lineage, which was associated with elevated signaling by innate immune mediators, such as IL-1ß and granulocyte-macrophage colony-stimulating factor (GM-CSF), and with adaptations in glucose metabolism and cholesterol biosynthesis. The trained-immunity-related increase in myelopoiesis resulted in a beneficial response to secondary LPS challenge and protection from chemotherapy-induced myelosuppression in mice. Therefore, modulation of myeloid progenitors in the bone marrow is an integral component of trained immunity, which to date, was considered to involve functional changes of mature myeloid cells in the periphery.


Assuntos
Imunidade Inata , Memória Imunológica , Células Progenitoras Mieloides/imunologia , Animais , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/efeitos dos fármacos , Mielopoese/imunologia , beta-Glucanas/farmacologia
2.
Nat Chem Biol ; 17(5): 567-575, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33664520

RESUMO

The discovery of effective therapeutic treatments for cancer via cell differentiation instead of antiproliferation remains a great challenge. Cyclin-dependent kinase 2 (CDK2) inactivation, which overcomes the differentiation arrest of acute myeloid leukemia (AML) cells, may be a promising method for AML treatment. However, there is no available selective CDK2 inhibitor. More importantly, the inhibition of only the enzymatic function of CDK2 would be insufficient to promote notable AML differentiation. To further validate the role and druggability of CDK2 involved in AML differentiation, a suitable chemical tool is needed. Therefore, we developed first-in-class CDK2-targeted proteolysis-targeting chimeras (PROTACs), which promoted rapid and potent CDK2 degradation in different cell lines without comparable degradation of other targets, and induced remarkable differentiation of AML cell lines and primary patient cells. These data clearly demonstrated the practicality and importance of PROTACs as alternative tools for verifying CDK2 protein functions.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Células Progenitoras Mieloides/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Triazóis/farmacologia , Antineoplásicos/síntese química , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Desenho de Fármacos , Descoberta de Drogas , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Concentração Inibidora 50 , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Células Progenitoras Mieloides/enzimologia , Células Progenitoras Mieloides/patologia , Piperazinas/farmacologia , Cultura Primária de Células , Piridinas/farmacologia , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais , Relação Estrutura-Atividade , Transcriptoma , Triazóis/síntese química
3.
PLoS Pathog ; 15(12): e1008115, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31887131

RESUMO

The opportunistic fungal pathogen Candida albicans can cause invasive infections in susceptible hosts and the innate immune system, in particular myeloid cell-mediated immunity, is critical for rapid immune protection and host survival during systemic candidiasis. Using a mouse model of the human disease, we identified a novel role of IL-23 in antifungal defense. IL-23-deficient mice are highly susceptible to systemic infection with C. albicans. We found that this results from a drastic reduction in all subsets of myeloid cells in the infected kidney, which in turn leads to rapid fungal overgrowth and renal tissue injury. The loss in myeloid cells is not due to a defect in emergency myelopoiesis or the recruitment of newly generated cells to the site of infection but, rather, is a consequence of impaired survival of myeloid cells at the site of infection. In fact, the absence of a functional IL-23 pathway causes massive myeloid cell apoptosis upon C. albicans infection. Importantly, IL-23 protects myeloid cells from apoptosis independently of the IL-23-IL-17 immune axis and independently of lymphocytes and innate lymphoid cells. Instead, our results suggest that IL-23 acts in a partially autocrine but not cell-intrinsic manner within the myeloid compartment to promote host protection from systemic candidiasis. Collectively, our data highlight an unprecedented and non-canonical role of IL-23 in securing survival of myeloid cells, which is key for maintaining sufficient numbers of cells at the site of infection to ensure efficient host protection.


Assuntos
Candida albicans/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Interleucina-23/farmacologia , Células Progenitoras Mieloides/efeitos dos fármacos , Animais , Candida albicans/imunologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo
4.
J Clin Pharm Ther ; 46(5): 1373-1381, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34101878

RESUMO

WHAT IS KNOWN AND OBJECTIVE: We have previously shown that the saponins of Sanguisorba parviflora (Maxim.) Takeda (Sp. T) relieved cyclophosphamide-induced myelosuppression in leukopenic mice. Haematopoietic cell-specific protein 1-associated protein X-1 (HAX-1) participated in the survival of neutrophils through the regulation of mitochondrial function. The aim of the present study was to comprehensively identify the role of HAX-1 in the mechanism of leukopenia alleviation by Sp. T. METHODS: HAX-1 gene and protein expression levels in peripheral blood neutrophils were examined using real-time quantitative reverse transcription-polymerase chain reaction, western blot and immunohistochemical assays. Neutrophil apoptosis was measured using flow cytometry. Mitochondrial function was determined via assessments of the reactive oxygen species (ROS) generation and mitochondrial membrane potential (ΔΨm) integrity levels. RESULTS AND DISCUSSION: The HAX-1 gene expression level in the peripheral blood neutrophils was significantly lower in patients with leukopenia than in healthy donors. The saponins of Sp. T induced HAX-1 expression and promoted myeloid progenitor cell (mEB8-ER cell) viability. HAX-1 overexpression reduced the production of ROS and maintained ΔΨm integrity. Cyclophosphamide-induced mitochondrial dysfunction and apoptosis could be abrogated by treatment with Sp. T or metformin. WHAT IS NEW AND CONCLUSION: Our data suggest a mechanism through which Sp. T protects against chemotherapy-induced leukopenia by regulating HAX-1 gene expression in a mitochondrial-dependent manner.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Ciclofosfamida/efeitos adversos , Leucopenia/induzido quimicamente , Leucopenia/tratamento farmacológico , Sanguisorba/química , Saponinas/farmacologia , Adulto , Apoptose/efeitos dos fármacos , Feminino , Humanos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Pessoa de Meia-Idade , Células Progenitoras Mieloides/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
5.
Invest New Drugs ; 38(3): 610-620, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31227936

RESUMO

The majority of patients with Philadelphia-negative myeloproliferative neoplasms (MPNs) harbor a gain of function mutation V617F in Janus kinase (JAK) 2. Although JAK2 inhibitors such as ruxolitinib have been shown to be clinically efficacious, the hematological toxicity and eventual drug resistance limit its use as monotherapy. Other gene mutations or dysregulation correlated with the disease phenotype and prognosis have been found to contribute to the complexity and heterogeneity of MPNs, giving rise to an increasing demand for combination therapies. Here, we combine ruxolitinib and the histone deacetylase inhibitor vorinostat as a rational combination strategy for MPNs. We tested the combination of ruxolitinib and vorinostat in cells with the JAK2V617F mutation, such as HEL cells, c-Kit+ cells from JAK2V617F transgenic mice and bone marrow mononuclear cells (BMMNCs) from patients with MPN. Our results showed significant synergistic effects of this combination strategy. Cotreatment with ruxolitinib and vorinostat synergistically induced apoptosis, cell cycle arrest and inhibition of the colony-forming capacity of HEL cells by attenuating the JAK/signal transducer and activator of transcription (STAT) and protein kinase-B (AKT) signaling pathways. In particular, cotreatment with ruxolitinib and vorinostat prevented the formation of large colonies of colony-forming unit-granulocyte/erythroid/macrophage/megakaryocytes (CFU-GEMMs) and colony-forming unit-granulocyte/macrophages (CFU-GMs) derived from the BMMNCs of patients with MPN. Taken together, these data provided preclinical evidence that the combination of ruxolitinib and vorinostat is a potential dual-target therapy for patients with MPN.


Assuntos
Histona Desacetilases/metabolismo , Janus Quinase 2/metabolismo , Transtornos Mieloproliferativos/tratamento farmacológico , Neoplasias/tratamento farmacológico , Pirazóis/farmacologia , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vorinostat/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Transgênicos , Mutação/genética , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Transtornos Mieloproliferativos/metabolismo , Neoplasias/metabolismo , Nitrilas , Pirimidinas
6.
J Transl Med ; 17(1): 247, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31366356

RESUMO

BACKGROUND: The molecular pathways that drive bone marrow myeloid progenitors (BMMP) development are very well understood and include a tight controlled multi-stage gene hierarch. Monocytes are versatile cells that display remarkable plasticity and may give rise to specific subsets of macrophages to proper promote tissue homesostasis upon an injury. However, the epigenetic mechanisms that underlie monocyte differentiation into the pro-inflammatory Ly6Chigh or the repairing Ly6Clow subsets are yet to be elucidated. We have previously shown that Epigenetic mechanisms Histone Deacetylase (HDAC) dependent are crucial for monocyte behavior and plasticity and in this work, we propose that this same mechanism underlies BMMP plasticity upon an inflammatory challenge in vivo. METHODS: BMMP were culture in the presence of GM-CSF alone or in combination with HDAC inhibitor (iHDAC) and phenotyped by flow cytometry, immune staining or western blot. iHDAC was topically added to skin wounds for 7 consecutive days and wound healing was monitored by flow cytometry and histopathological analysis. RESULTS: When BMMP were cultured in the presence of iHDAC, we showed that the CD11blow/Ly6Clow subset was the specific target of iHDAC that underwent chromatin hyperacetylation in vitro. Upon 13 days in the presence of iHDAC, BMMP gave rise to very elongated macrophages, that in turn, displayed a remarkable plasticity in a HDAC activity dependent fashion. HDAC-dependent cell shape was tight related to macrophage behavior and phenotype through the control of iNOS protein levels, showing that chromatin remodeling is a key component of macrophage plasticity and function. We then hypothesized that iHDAC would modulate the inflammatory response and favor tissue repair in vivo. To test this hypothesis, we topically added iHDAC to skin wounds during 7 consecutive days and followed tissue repair dynamics. In fact, iHDAC treated skin wounds presented an increase in wound closure at day 5 that was correlated to an enrichment in the CD11blow/Ly6Clow subset and in very elongated F4/80 positives macrophages in vivo, fully recapitulating the behavior previously observed in vitro. CONCLUSION: Our work provides the biological basis that connects chromatin remodeling to phenotypic plasticity, which in turn, may become a tractable therapeutic strategy in further translational studies.


Assuntos
Epigenoma , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Células Progenitoras Mieloides/citologia , Pele/efeitos dos fármacos , Pele/patologia , Cicatrização , Animais , Cromatina/química , Epigênese Genética , Histona Desacetilases/genética , Humanos , Inflamação , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Fenótipo
7.
Immunol Invest ; 48(1): 96-105, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30204524

RESUMO

Previously, we found that dual therapy by the CXCR4 inhibitor Plerixafor and cytosine arabinoside (Ara-C) effectively eradicated leukemia cells and concurrently activated immune cells in acute myeloid leukemia (AML). To reveal the significance of programmed death-ligand1 (PD-L1) in AML and as a strategic approach, we investigated the anti-leukemic effect of a triple combinational therapy by utilizing Plerixafor and anti-PD-L1 in combination with chemotherapy in an AML mouse model. We examined leukemic myeloid blast cells in multiple organs after the successive treatment with Ara-C, Plerixafor, and anti-PD-L1. The results showed that noticeable benefits of triple combinational therapy for eradication of myeloid blast cells in vivo with prolonged survival rates. The frequencies of regulatory T cells (Tregs), monocytic-myeloid-derived suppressor cells (M-MDSCs), and granulocytic-myeloid-derived suppressor cells (G-MDSCs), in the peripheral blood of leukemic mice were consistently decreased, even when mice were sacrificed alive at D + 26 after completion of the triple combinational therapy, compared to the other subgroups. These findings imply that the modulation by the triple combinational therapy may lead to more efficient leukemic myeloid blast cell ablation through the suppression of Tregs or M-MDSCs and G-MDSCs in AML. Although Plerixafor and PD-L1 antagonist do not have a direct anti-leukemic role, our results provide some clues and guidelines to develop clinically therapeutic strategies for chemotherapy-resistant patients by the modulation of leukemic microenvironments.


Assuntos
Antígeno B7-H1/imunologia , Imunoterapia/métodos , Leucemia Mieloide Aguda/terapia , Células Progenitoras Mieloides/efeitos dos fármacos , Receptores CXCR4/antagonistas & inibidores , Animais , Anticorpos Monoclonais/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzilaminas , Linhagem Celular Tumoral , Ciclamos , Citarabina/uso terapêutico , Modelos Animais de Doenças , Compostos Heterocíclicos/uso terapêutico , Humanos , Imunomodulação , Leucemia Mieloide Aguda/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/fisiologia , Microambiente Tumoral/efeitos dos fármacos
8.
J Biol Chem ; 291(7): 3439-54, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26670608

RESUMO

Osteoclasts are specialized polyploid cells that resorb bone. Upon stimulation with receptor activator of nuclear factor-κB ligand (RANKL), myeloid precursors commit to becoming polyploid, largely via cell fusion. Polyploidization of osteoclasts is necessary for their bone-resorbing activity, but the mechanisms by which polyploidization is controlled remain to be determined. Here, we demonstrated that in addition to cell fusion, incomplete cytokinesis also plays a role in osteoclast polyploidization. In in vitro cultured osteoclasts derived from mice expressing the fluorescent ubiquitin-based cell cycle indicator (Fucci), RANKL induced polyploidy by incomplete cytokinesis as well as cell fusion. Polyploid cells generated by incomplete cytokinesis had the potential to subsequently undergo cell fusion. Nuclear polyploidy was also observed in osteoclasts in vivo, suggesting the involvement of incomplete cytokinesis in physiological polyploidization. Furthermore, RANKL-induced incomplete cytokinesis was reduced by inhibition of Akt, resulting in impaired multinucleated osteoclast formation. Taken together, these results reveal that RANKL-induced incomplete cytokinesis contributes to polyploidization of osteoclasts via Akt activation.


Assuntos
Núcleo Celular/metabolismo , Citocinese , Células Progenitoras Mieloides/metabolismo , Osteoclastos/metabolismo , Osteólise/metabolismo , Poliploidia , Ligante RANK/metabolismo , Animais , Benzimidazóis/farmacologia , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Fusão Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Células Cultivadas , Cruzamentos Genéticos , Citocinese/efeitos dos fármacos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos Transgênicos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/patologia , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteólise/patologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/agonistas , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinoxalinas/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
9.
Stem Cells ; 34(7): 1859-71, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26972179

RESUMO

Previous studies indicate that Kras is dispensable for fetal liver hematopoiesis, but its role in adult hematopoiesis remains unclear. Here, we generated a Kras conditional knockout allele to address this question. Deletion of Kras in adult bone marrow (BM) is mediated by Vav-Cre or inducible Mx1-Cre. We find that loss of Kras leads to greatly reduced thrombopoietin (TPO) signaling in hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs), while stem cell factor-evoked ERK1/2 activation is not affected. The compromised TPO signaling is associated with reduced long term- and intermediate-term HSC compartments and a bias toward myeloid differentiation in MPPs. Although granulocyte macrophage colony-stimulating factor (GM-CSF)-evoked ERK1/2 activation is only moderately decreased in Kras(-/-) myeloid progenitors, it is blunted in neutrophils and neutrophil survival is significantly reduced in vitro. At 9-12 months old, Kras conditional knockout mice develop profound hematopoietic defects, including splenomegaly, an expanded neutrophil compartment, and reduced B cell number. In a serial transplantation assay, the reconstitution potential of Kras(-/-) BM cells is greatly compromised, which is attributable to defects in the self-renewal of Kras(-/-) HSCs and defects in differentiated hematopoietic cells. Our results demonstrate that Kras is a major regulator of TPO and GM-CSF signaling in specific populations of hematopoietic cells and its function is required for adult hematopoiesis. Stem Cells 2016;34:1859-1871.


Assuntos
Envelhecimento/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Compartimento Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Deleção de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/deficiência
10.
Carcinogenesis ; 37(7): 692-700, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207665

RESUMO

Benzene, formaldehyde (FA) and trichloroethylene (TCE) are ubiquitous chemicals in workplaces and the general environment. Benzene is an established myeloid leukemogen and probable lymphomagen. FA is classified as a myeloid leukemogen but has not been associated with non-Hodgkin lymphoma (NHL), whereas TCE has been associated with NHL but not myeloid leukemia. Epidemiologic associations between FA and myeloid leukemia, and between benzene, TCE and NHL are, however, still debated. Previously, we showed that these chemicals are associated with hematotoxicity in cross-sectional studies of factory workers in China, which included extensive personal monitoring and biological sample collection. Here, we compare and contrast patterns of hematotoxicity, monosomy 7 in myeloid progenitor cells (MPCs), and B-cell activation biomarkers across these studies to further evaluate possible mechanisms of action and consistency of effects with observed hematologic cancer risks. Workers exposed to benzene or FA, but not TCE, showed declines in cell types derived from MPCs, including granulocytes and platelets. Alterations in lymphoid cell types, including B cells and CD4+ T cells, and B-cell activation markers were apparent in workers exposed to benzene or TCE. Given that alterations in myeloid and lymphoid cell types are associated with hematological malignancies, our data provide biologic insight into the epidemiological evidence linking benzene and FA exposure with myeloid leukemia risk, and TCE and benzene exposure with NHL risk.


Assuntos
Benzeno/toxicidade , Formaldeído/toxicidade , Leucemia/induzido quimicamente , Linfoma não Hodgkin/induzido quimicamente , Tricloroetileno/toxicidade , Adulto , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Biomarcadores Tumorais/metabolismo , China , Feminino , Hemolíticos/toxicidade , Humanos , Leucemia/epidemiologia , Leucemia/patologia , Ativação Linfocitária/efeitos dos fármacos , Linfoma não Hodgkin/epidemiologia , Linfoma não Hodgkin/patologia , Masculino , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/patologia , Exposição Ocupacional
11.
Blood ; 124(3): 393-402, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24894772

RESUMO

Bone morphogenetic protein (BMP) signaling regulates early hematopoietic development, proceeding from mesoderm patterning through the progressive commitment and differentiation of progenitor cells. The BMP pathway signals largely through receptor-mediated activation of Mothers Against Decapentaplegic homolog (SMAD) proteins, although alternate pathways are modulated through various components of mitogen-activated protein kinase (MAPK) signaling. Using a conditional, short hairpin RNA (shRNA)-based knockdown system in the context of differentiating embryonic stem cells (ESCs), we demonstrated previously that Smad1 promotes hemangioblast specification, but then subsequently restricts primitive progenitor potential. Here we show that co-knockdown of Smad5 restores normal progenitor potential of Smad1-depleted cells, suggesting opposing functions for Smad1 and Smad5. This balance was confirmed by cotargeting Smad1/5 with a specific chemical antagonist, LDN193189 (LDN). However, we discovered that LDN treatment after hemangioblast commitment enhanced primitive myeloid potential. Moreover, inhibition with LDN (but not SMAD depletion) increased expression of Delta-like ligands Dll1 and Dll3 and NOTCH activity; abrogation of NOTCH activity restored LDN-enhanced myeloid potential back to normal, corresponding with expression levels of the myeloid master regulator, C/EBPα. LDN but not SMAD activity was also associated with activation of the p38MAPK pathway, and blocking this pathway was sufficient to enhance myelopoiesis. Therefore, NOTCH and p38MAPK pathways balance primitive myeloid progenitor output downstream of the BMP pathway.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Mielopoese/fisiologia , Receptores Notch/metabolismo , Proteínas Smad/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Corpos Embrioides/citologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Mielopoese/efeitos dos fármacos , Mielopoese/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Proteína Smad5/metabolismo
12.
BMC Cancer ; 16: 440, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27400708

RESUMO

BACKGROUND: Previous work has demonstrated immunomodulatory, anti-tumor, anti-metastatic and anti-angiogenic effects of the small molecule quinoline-3-carboxamide tasquinimod in pre-clinical cancer models. To better understand the anti-tumor effects of tasquinimod in transplantable tumor models, we have evaluated the impact of the compound both on recruitment of myeloid cells to tumor tissue and on tumor-induced myeloid cell expansion as these cells are known to promote tumor development. METHODS: Mice bearing subcutaneous 4 T1 mammary carcinoma tumors were treated with tasquinimod in the drinking water. A BrdU-based flow cytometry assay was utilized to assess the impact of short-term tasquinimod treatment on myeloid cell recruitment to tumors. Additionally, long-term treatment was performed to study the anti-tumor effect of tasquinimod as well as its effects on splenic myeloid cells and their progenitors. Myeloid cell populations were also immune-depleted by in vivo antibody treatment. RESULTS: Short-term tasquinimod treatment did not influence the proliferation of splenic Ly6C(hi) and Ly6G(hi) cells, but instead reduced the influx of Ly6C(hi) cells to the tumor. Treatment with tasquinimod for various periods of time after tumor inoculation revealed that the anti-tumor effect of this compound mainly operated during the first few days of tumor growth. Similar to tasquinimod treatment, antibody-mediated depletion of Ly6C(hi) cells within that same time frame, caused reduced tumor growth, thereby confirming a significant role for these cells in tumor development. Additionally, long-term tasquinimod treatment reduced the splenomegaly and expansion of splenic myeloid cells during a later phase of tumor development. In this phase, tasquinimod normalized the tumor-induced alterations in myeloerythroid progenitor cells in the spleen but had only limited impact on the same populations in the bone marrow. CONCLUSIONS: Our results indicate that tasquinimod treatment reduces tumor growth by operating early after tumor inoculation and that this effect is at least partially caused by reduced recruitment of Ly6C(hi) cells to tumor tissue. Long-term treatment also reduces the number of splenic myeloid cells and myeloerythroid progenitors, but these effects did not influence established rapidly growing tumors.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Quinolinas/farmacologia , Quinolonas/farmacologia , Administração Oral , Inibidores da Angiogênese/administração & dosagem , Animais , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Células Mieloides/patologia , Células Progenitoras Mieloides/efeitos dos fármacos , Mielopoese/efeitos dos fármacos , Quinolinas/administração & dosagem , Quinolonas/administração & dosagem , Baço/citologia , Baço/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Brain Behav Immun ; 57: 293-303, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27223094

RESUMO

In order to relieve anxiety and depression accompanying stress, physicians resort to tricyclic antidepressants, such as imipramine. We had previously shown that imipramine reversed stress-induced social avoidance behavior, and down-regulated microglial activation 24days after stress cessation. To further characterize the effects of imipramine on stress induced neuroimmune dysregulation and associated changes in behavior, the aims of this study were to determine if imipramine 1) ameliorated stress-induced inflammation in the periphery and central nervous system, and 2) prevented stress related anxiety- and depressive-like behaviors. C57BL/6 mice were treated with imipramine (15mg/kg) in their drinking water, and exposed to repeated social defeat (RSD). Imipramine attenuated stress-induced corticosterone and IL-6 responses in plasma. Imipramine decreased the percentage of monocytes and granulocytes in the bone marrow and circulation. However, imipramine did not prevent splenomegaly, stress-related increased percentage of granulocytes in this organ, and the production of pro-inflammatory cytokines in the spleen, following RSD. Moreover, imipramine abrogated the accumulation of macrophages in the brain in mice exposed to RSD. Imipramine blocked neuroinflammatory signaling and prevented stress-related anxiety- and depressive-like behaviors. These data support the notion that pharmacomodulation of the monoaminergic system, besides exerting anxiolytic and antidepressant effects, may have therapeutic effects as a neuroimmunomodulator during stress.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Ansiedade , Corticosterona/sangue , Depressão , Imipramina/farmacologia , Inflamação , Interleucina-6/sangue , Células Progenitoras Mieloides/efeitos dos fármacos , Baço , Estresse Psicológico , Animais , Antidepressivos Tricíclicos/administração & dosagem , Ansiedade/etiologia , Ansiedade/imunologia , Ansiedade/metabolismo , Ansiedade/prevenção & controle , Comportamento Animal , Depressão/etiologia , Depressão/imunologia , Depressão/metabolismo , Depressão/prevenção & controle , Imipramina/administração & dosagem , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/imunologia , Estresse Psicológico/metabolismo
14.
Carcinogenesis ; 36(1): 160-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25391402

RESUMO

Formaldehyde (FA) is an economically important industrial chemical to which millions of people worldwide are exposed environmentally and occupationally. Recently, the International Agency for Cancer Research concluded that there is sufficient evidence that FA causes leukemia, particularly myeloid leukemia. To evaluate the biological plausibility of this association, we employed a chromosome-wide aneuploidy study approach, which allows the evaluation of aneuploidy and structural chromosome aberrations (SCAs) of all 24 chromosomes simultaneously, to analyze cultured myeloid progenitor cells from 29 workers exposed to relatively high levels of FA and 23 unexposed controls. We found statistically significant increases in the frequencies of monosomy, trisomy, tetrasomy and SCAs of multiple chromosomes in exposed workers compared with controls, with particularly notable effects for monosomy 1 [P = 6.02E-06, incidence rate ratio (IRR) = 2.31], monosomy 5 (P = 9.01E-06; IRR = 2.24), monosomy 7 (P = 1.57E-05; IRR = 2.17), trisomy 5 (P = 1.98E-05; IRR = 3.40) and SCAs of chromosome 5 (P = 0.024; IRR = 4.15). The detection of increased levels of monosomy 7 and SCAs of chromosome 5 is particularly relevant as they are frequently observed in acute myeloid leukemia. Our findings provide further evidence that leukemia-related cytogenetic changes can occur in the circulating myeloid progenitor cells of healthy workers exposed to FA, which may be a potential mechanism underlying FA-induced leukemogenesis.


Assuntos
Aneuploidia , Cromossomos Humanos/efeitos dos fármacos , Desinfetantes/efeitos adversos , Formaldeído/efeitos adversos , Células Progenitoras Mieloides/efeitos dos fármacos , Exposição Ocupacional/efeitos adversos , Adulto , Estudos de Casos e Controles , Células Cultivadas , Estudos Transversais , Feminino , Seguimentos , Humanos , Hibridização in Situ Fluorescente , Masculino , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Prognóstico
15.
Carcinogenesis ; 36(5): 585-97, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25784375

RESUMO

Snail2 is a zinc finger transcription factor involved in driving epithelial to mesenchymal transitions. Snail2 null mice are viable, but display defects in melanogenesis, gametogenesis and hematopoiesis, and are markedly radiosensitive. Here, using mouse genetics, we have studied the contributions of Snail2 to epidermal homeostasis and skin carcinogenesis. Snail2 (-/-) mice presented a defective epidermal terminal differentiation and, unexpectedly, an increase in number, size and malignancy of tumor lesions when subjected to the two-stage mouse skin chemical carcinogenesis protocol, compared with controls. Additionally, tumor lesions from Snail2 (-/-) mice presented a high inflammatory component with an elevated percentage of myeloid precursors in tumor lesions that was further increased in the presence of the anti-inflammatory agent dexamethasone. In vitro studies in Snail2 null keratinocytes showed that loss of Snail2 leads to a decrease in proliferation indicating a non-cell autonomous role for Snail2 in the skin carcinogenic response observed in vivo. Bone marrow (BM) cross-reconstitution assays between Snail2 wild-type and null mice showed that Snail2 absence in the hematopoietic system fully reproduces the tumor behavior of the Snail2 null mice and triggers the accumulation of myeloid precursors in the BM, blood and tumor lesions. These results indicate a new role for Snail2 in preventing myeloid precursors recruitment impairing skin chemical carcinogenesis progression.


Assuntos
Inflamação/patologia , Queratinócitos/patologia , Células Progenitoras Mieloides/patologia , Neoplasias Experimentais/patologia , Neoplasias Cutâneas/patologia , Fatores de Transcrição/fisiologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Apoptose , Western Blotting , Carcinógenos/toxicidade , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Imunofluorescência , Hematopoese , Técnicas Imunoenzimáticas , Inflamação/induzido quimicamente , Inflamação/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição da Família Snail
16.
Immunopharmacol Immunotoxicol ; 37(4): 359-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26153910

RESUMO

Increased numbers of myeloid-derived suppressor cells (MDSCs) are often observed in various pathological and physiological conditions. However, the interactions between neurotransmitters and MDSCs have not been elucidated. In this study, we studied whether norepinephrine (NE), a neurotransmitter, could affect the differentiation of human MDSCs in vitro. Flow cytometric analysis showed that treatment with 20 µM NE significantly enhanced the expansion of MDSCs. The NE-generated MDSCs suppressed the T-cells proliferation, depending on the production of reactive oxygen species (ROS). Moreover, the expansion of MDSCs induced by NE resulted in a dramatic induction of nicotinamide adenine dinucleotide phosphate oxidase subunit P47(phox). Addition of the ROS inhibitor catalase into the MDSCs/T-cell co-culture system partly abrogated the suppressive effects of MDSCs on T-cell proliferation. In summary, our data have shown that NE enhanced the expansion of human MDSCs in vitro, providing important insights into the novel roles of neurotransmitters in the regulation of myeloid cell differentiation and function.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Neurotransmissores/farmacologia , Norepinefrina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/efeitos dos fármacos , Diferenciação Celular/imunologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Células Mieloides/citologia , Células Mieloides/metabolismo , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/metabolismo , Óxido Nítrico/biossíntese , Linfócitos T/citologia , Linfócitos T/metabolismo
17.
Br J Cancer ; 111(7): 1350-62, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25101565

RESUMO

BACKGROUND: Host immunity is emerging as a key player in the prognosis and response to treatment of cancer patients. However, the impact of the immune system and its modulation by therapies are unknown in rare soft tissue sarcomas such as solitary fibrous tumours (SFTs), whose management in the advanced forms includes anti-angiogenic therapy. Here, we studied the in situ and systemic immune status of advanced SFT patients and the effects of sunitinib malate (SM) in association with the clinical efficacy. METHODS: Immune contexture of SFTs was assessed by immunohistochemistry in lesions from untreated or SM-treated patients. Frequency of circulating myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and T-cell functions was assessed ex vivo in SFT patients prior and during anti-angiogenic therapy. Patients with long-term tumour control were included to correlate immune profiles and clinical responses. RESULTS: Anti-angiogenic naïve SFT lesions were heavily infiltrated by CD163(+)CD14(+)CD68(-) and CD163(+)CD14(-)CD68(-) myeloid cells but devoid of T cells. Conversely, post-SM tumours acquired a new subset of CD68(+)CD14(+) myeloid cells and displayed traits of an on-going adaptive immunity, strongly enriched in activated CD8(+) and CD4(+) T cells. These changes at the tumour site paralleled the alleviation of systemic immunosuppression and the drop in the frequency of circulating monocytic MDSCs (mMDSCs) and granulocytic MDSCs (gMDSCs). Rebound in the number of mMDSCs, but not of gMDSCs occurred at disease progression, and a reduced percentages of mMDSCs, comparable to those found in healthy donors (HDs), endured only in the SM-responsive patients. CONCLUSIONS: The immune contexture of SFT patients is heavily involved in anti-angiogenic therapy and it could be exploited to achieve more durable disease control through immune-based combination strategies.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Indóis/farmacologia , Células Progenitoras Mieloides/imunologia , Pirróis/farmacologia , Tumores Fibrosos Solitários/imunologia , Adulto , Idoso , Inibidores da Angiogênese/uso terapêutico , Intervalo Livre de Doença , Feminino , Humanos , Terapia de Imunossupressão , Indóis/uso terapêutico , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Masculino , Pessoa de Meia-Idade , Células Progenitoras Mieloides/efeitos dos fármacos , Pirróis/uso terapêutico , Tumores Fibrosos Solitários/sangue , Tumores Fibrosos Solitários/tratamento farmacológico , Sunitinibe , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
18.
Blood Cells Mol Dis ; 50(3): 171-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23265740

RESUMO

Elucidating ways to enhance megakaryopoiesis in vivo would have therapeutic applications for thrombocytopenia and transfusion medicine. Nicotinamide has been shown to enhance endomitosis in megakaryocytes cultured in vitro, suggesting that it may be beneficial for the production of platelets in culture. We hypothesized that regular injections of nicotinamide in mice would also increase platelets in vivo. However, we found that platelet counts were reduced by about 25% with daily injections of nicotinamide. Altering the schedule, duration, or nicotinamide dose did not improve platelet production. Consistent with lower platelet levels, nicotinamide also tended to decrease megakaryocyte frequency in sternum and spleen sections, as well as colony formation in vitro by bone marrow progenitor cells. However, there was no effect on the fraction or ploidy of CD41(+) cells harvested from bone marrow. Together, our results suggest that, although nicotinamide increases polyploidization of megakaryocytes in culture, it does not have translatable effects in vivo.


Assuntos
Plaquetas/efeitos dos fármacos , Niacinamida/administração & dosagem , Contagem de Plaquetas , Animais , Células da Medula Óssea/citologia , Feminino , Masculino , Megacariócitos/citologia , Camundongos , Células Progenitoras Mieloides/efeitos dos fármacos , Baço/citologia , Trombopoese/efeitos dos fármacos
19.
J Virol ; 86(18): 9854-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761372

RESUMO

Human cytomegalovirus (HCMV) is a herpesvirus that establishes a lifelong, latent infection within a host. At times when the immune system is compromised, the virus undergoes a lytic reactivation producing infectious progeny. The identification and understanding of the biological mechanisms underlying HCMV latency and reactivation are not completely defined. To this end, we have developed a tractable in vitro model system to investigate these phases of viral infection using a clonal population of myeloid progenitor cells (Kasumi-3 cells). Infection of these cells results in maintenance of the viral genome with restricted viral RNA expression that is reversed with the addition of the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA, also known as PMA). Additionally, a latent viral transcript (LUNA) is expressed at times where viral lytic transcription is suppressed. Infected Kasumi-3 cells initiate production of infectious virus following TPA treatment, which requires cell-to-cell contact for efficient transfer of virus to other cell types. Importantly, lytically infected fibroblast, endothelial, or epithelial cells can transfer virus to Kasumi-3 cells, which fail to initiate lytic replication until stimulated with TPA. Finally, inflammatory cytokines, in addition to the pharmacological agent TPA, are sufficient for transcription of immediate-early (IE) genes following latent infection. Taken together, our findings argue that the Kasumi-3 cell line is a tractable in vitro model system with which to study HCMV latency and reactivation.


Assuntos
Citomegalovirus/fisiologia , Citomegalovirus/patogenicidade , Células Progenitoras Mieloides/virologia , Sequência de Bases , Linhagem Celular , Citocinas/fisiologia , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , DNA Viral/genética , Expressão Gênica/efeitos dos fármacos , Genes Precoces , Genoma Viral , Humanos , Mediadores da Inflamação/fisiologia , Modelos Biológicos , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Ativação Viral/genética , Ativação Viral/fisiologia , Latência Viral/genética , Latência Viral/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
20.
Blood ; 117(13): 3548-58, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21173117

RESUMO

Because lymphoid progenitors can give rise to natural killer (NK) cells, NK ontogeny has been considered to be exclusively lymphoid. Here, we show that rare human CD34(+) hematopoietic progenitors develop into NK cells in vitro in the presence of cytokines (interleukin-7, interleukin-15, stem cell factor, and fms-like tyrosine kinase-3 ligand). Adding hydrocortisone and stromal cells greatly increases the frequency of progenitor cells that give rise to NK cells through the recruitment of myeloid precursors, including common myeloid progenitors and granulocytic-monocytic precursors to the NK-cell lineage. WNT signaling was involved in this effect. Cells at more advanced stages of myeloid differentiation (with increasing expression of CD13 and macrophage colony-stimulating factor receptor [M-CSFR]) could also differentiate into NK cells in the presence of cytokines, stroma, and hydrocortisone. NK cells derived from myeloid precursors (CD56(-)CD117(+)M-CSFR(+)) showed more expression of killer immunoglobulin-like receptors, a fraction of killer immunoglobulin-like receptor-positive-expressing cells that lacked NKG2A, a higher cytotoxicity compared with CD56(-)CD117(+)M-CSFR(-) precursor-derived NK cells and thus resemble the CD56(dim) subset of NK cells. Collectively, these studies show that NK cells can be derived from the myeloid lineage.


Assuntos
Diferenciação Celular/fisiologia , Células Matadoras Naturais/fisiologia , Células Progenitoras Mieloides/fisiologia , Antígenos CD34/metabolismo , Antígeno CD56/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Humanos , Hidrocortisona/farmacologia , Células K562 , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA