Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Int J Cancer ; 152(12): 2615-2628, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36912275

RESUMO

Due to a combination of rapid disease progression and the lack of curative treatment options, hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Infiltrated, monocyte-derived, tumor-associated macrophages are known to play a role in HCC pathogenesis, but the involvement of Kupffer cells (KCs) remains elusive. Here, we used the Clec4F-diphteria toxin receptor transgenic mouse model to specifically investigate the effect of KC depletion on HCC initiation, progression and neoplastic growth following liver resection. For this purpose, several HCC mouse models with varying underlying etiologies were used and partial hepatectomy was performed. Our results show that in HCC, developed on a fibrotic or non-alcoholic steatohepatitis background, depletion of embryonic KCs at the onset of HCC induction and the subsequent replacement by monocyte-derived KCs does not affect the tumor burden, tumor microenvironment or the phenotype of isolated KCs at end-stage disease. In non-chronic liver disease-associated diethylnitrosamine-induced HCC, ablation of Clec4F+ KCs did not alter tumor progression or neoplastic growth following liver resection. Our results show that temporal ablation of resident KCs does not impact HCC pathogenesis, neither in the induction phase nor in advanced disease, and indicate that bone marrow-derived KCs are able to swiftly repopulate the available KC niche and adopt their phenotype.


Assuntos
Carcinogênese , Carcinoma Hepatocelular , Células de Kupffer , Neoplasias Hepáticas Experimentais , Neoplasias Hepáticas , Macrófagos Associados a Tumor , Células de Kupffer/imunologia , Progressão da Doença , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/patologia , Animais , Camundongos , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/patologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Carcinogênese/imunologia , Carcinogênese/patologia , Camundongos Endogâmicos C57BL , Masculino
2.
Am J Surg Pathol ; 45(10): 1391-1398, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34172624

RESUMO

Blast evaluation in patients with acute monocytic leukemias (AMoL) is notoriously difficult due to the lack of reliable surface markers and cytologic subtleties on the aspirate smears. While blasts of most nonmonocytic acute leukemias express CD34, available immunohistochemical antibodies to monocytic blasts also mark normal background mature monocytes. We searched for a potential biomarker candidate by surveying specific gene expression profiles of monocyte progenitors. Our investigations led us to IRF8, which is a lineage-specific transcription factor critical for the production of monocytic and dendritic cell progenitors. In this study, we tested and validated a monoclonal antibody to IRF8 as a novel immunohistochemical stain for trephine core biopsies of human bone marrow. We assessed the expression of IRF8 in 90 cases of AMoL, including posttherapy staging bone marrows, 23 cases of chronic myelomonocytic leukemia, 26 cases of other acute myeloid leukemia subtypes, and 18 normal control marrows. In AMoL, there was high correlation of IRF8-positive cells to aspirate blast count (R=0.95). Comparison of IRF8 staining to aspirate blast percentage in chronic myelomonocytic leukemia also showed good correlation (R=0.86). In contrast, IRF8-positive cells did not correlate with blast count in other subtypes of acute myeloid leukemia (R=0.56) and staining was <5% in all normal control marrows, even those with reactive monocytosis. We found that IRF8 was also weakly reactive in B cells and hematogones, with the latter accounting for rare cases of discrepancies. When IRF8 was used to categorize cases as AMoL, positive for residual leukemia or negative, the sensitivity was 98%, specificity was 82%, positive predictive value was 86%, and negative predictive value was 98%. These results demonstrate that IRF8 may serve as a clinically useful immunostain to diagnose and track AMoLs on bone marrow core biopsies. This can be particularly impactful in the setting of poor aspiration and focal blast increase. In the era of new targeted therapies that have been reported to induce monocytic outgrowths of leukemia, a marker for malignant monoblasts may prove even more critical.


Assuntos
Biomarcadores Tumorais/análise , Imuno-Histoquímica , Fatores Reguladores de Interferon/análise , Leucemia Monocítica Aguda/metabolismo , Células Precursoras de Monócitos e Macrófagos/química , Idoso , Biópsia , Exame de Medula Óssea , Feminino , Humanos , Leucemia Monocítica Aguda/imunologia , Leucemia Monocítica Aguda/patologia , Masculino , Pessoa de Meia-Idade , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/patologia , Valor Preditivo dos Testes , Estudo de Prova de Conceito , Reprodutibilidade dos Testes
3.
Clin Immunol ; 201: 4-14, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30771501

RESUMO

Bacillus Calmette-Guerin (BCG) is one of the most effective treatments for bladder cancer. Little attention has been paid to the possible role of neutrophils in BCG immunotherapy. In this study, we examined neutrophil extracellular traps (NETs) formation induced by BCG stimulation, and found that BCG-induced NETs exerted cytotoxicity, induced apoptosis and cell-cycle arrest, and inhibited migration in bladder tumor cells. BCG-activated tumor cells but not non-activated ones elicited NETs formation, in which IL-8 and TNF-α from activated tumor cells both took effect. Moreover, NETs activated peripheral blood mononuclear cells (PBMCs) exhibited a higher expression of CD4 and Th1 cytokines. Additionally, the role of NETs in vivo contributed to the recruitment of T cells and monocytes-macrophages and tissue damage, thus preventing tumor growth. NETs proteins mainly caused these effects on tumor and cellular immunity. In conclusion, we demonstrated a novel immunoregulatory role for NETs in the early stages of BCG immunotherapy.


Assuntos
Armadilhas Extracelulares/imunologia , Mycobacterium bovis , Neutrófilos/imunologia , Neoplasias da Bexiga Urinária/terapia , Animais , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Citocinas/imunologia , Humanos , Imunoterapia , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/imunologia , Linfócitos T/imunologia , Neoplasias da Bexiga Urinária/imunologia
4.
Cell ; 175(4): 1014-1030.e19, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343900

RESUMO

Although current immune-checkpoint therapy (ICT) mainly targets lymphoid cells, it is associated with a broader remodeling of the tumor micro-environment. Here, using complementary forms of high-dimensional profiling, we define differences across all hematopoietic cells from syngeneic mouse tumors during unrestrained tumor growth or effective ICT. Unbiased assessment of gene expression of tumor-infiltrating cells by single-cell RNA sequencing (scRNAseq) and longitudinal assessment of cellular protein expression by mass cytometry (CyTOF) revealed significant remodeling of both the lymphoid and myeloid intratumoral compartments. Surprisingly, we observed multiple subpopulations of monocytes/macrophages, distinguishable by the markers CD206, CX3CR1, CD1d, and iNOS, that change over time during ICT in a manner partially dependent on IFNγ. Our data support the hypothesis that this macrophage polarization/activation results from effects on circulatory monocytes and early macrophages entering tumors, rather than on pre-polarized mature intratumoral macrophages.


Assuntos
Linfócitos/imunologia , Células Mieloides/imunologia , Neoplasias/imunologia , Análise de Célula Única , Transcriptoma , Animais , Linhagem Celular Tumoral , Citometria de Fluxo , Imunoterapia/métodos , Interferon gama/imunologia , Ativação de Macrófagos , Masculino , Espectrometria de Massas , Camundongos , Células Precursoras de Monócitos e Macrófagos/imunologia , Neoplasias/terapia
5.
FEBS J ; 284(15): 2410-2424, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28605567

RESUMO

Myeloid-derived suppressor cells (MDSCs), including granulocytic (G)-MDSCs and monocytic (M)-MDSCs, play a critical role in tumor-induced T cell tolerance. MDSC immunosuppressive function and differentiation are significantly promoted in patients and B-cell lymphoma model mice. However, the mechanisms regulating these processes remain largely unclear. In the present study, we observed increased microRNA (miR)-30a expression both in G-MDSCs and in M-MDSCs from B cell lymphoma model mice. After transfection with miR-30a mimics, the differentiation and suppressive capacities of MDSCs were significantly increased via up-regulation of arginase-1. Moreover, we showed that the 3'-UTR of suppressor of cytokine signaling 3 (SOCS3) mRNA is a direct target of miR-30a. Decreased SOCS3 expression and activated Janus kinase-signal transducer and activator of transcription 3 signaling promote MDSC differentiation and suppressive activities. These findings provide new insights into the molecular mechanisms underlying MDSC expansion and function during B cell lymphoma development.


Assuntos
Regiões 3' não Traduzidas , Diferenciação Celular , Linfoma de Células B/metabolismo , MicroRNAs/metabolismo , Células Supressoras Mieloides/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Regulação para Cima , Animais , Arginase/genética , Arginase/metabolismo , Células da Medula Óssea/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linhagem Celular Tumoral , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Células Precursoras de Granulócitos/imunologia , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/patologia , Terapia de Imunossupressão , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Linfoma de Células B/terapia , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/antagonistas & inibidores , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Células Precursoras de Monócitos e Macrófagos/patologia , Células Supressoras Mieloides/citologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Baço/patologia , Proteína 3 Supressora da Sinalização de Citocinas/genética
6.
Sci Rep ; 7: 40814, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28094319

RESUMO

Helminths cause chronic infections and affect the immune response to unrelated inflammatory diseases. Although helminths have been used therapeutically to ameliorate inflammatory conditions, their anti-inflammatory properties are poorly understood. Alternatively activated macrophages (AAMϕs) have been suggested as the anti-inflammatory effector cells during helminth infections. Here, we define the origin of AAMϕs during infection with Taenia crassiceps, and their disease-modulating activity on the Experimental Autoimmune Encephalomyelitis (EAE). Our data show two distinct populations of AAMϕs, based on the expression of PD-L1 and PD-L2 molecules, resulting upon T. crassiceps infection. Adoptive transfer of Ly6C+ monocytes gave rise to PD-L1+/PD-L2+, but not PD-L1+/PD-L2- cells in T. crassiceps-infected mice, demonstrating that the PD-L1+/PD-L2+ subpopulation of AAMϕs originates from blood monocytes. Furthermore, adoptive transfer of PD-L1+/PD-L2+ AAMϕs into EAE induced mice reduced disease incidence, delayed disease onset, and diminished the clinical disability, indicating the critical role of these cells in the regulation of autoimmune disorders.


Assuntos
Transferência Adotiva/métodos , Antígenos Ly/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Ativação de Macrófagos , Células Precursoras de Monócitos e Macrófagos/imunologia , Taenia/imunologia , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Células Cultivadas , Encefalomielite Autoimune Experimental/terapia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 2 Ligante de Morte Celular Programada 1/genética , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo
7.
PLoS One ; 11(5): e0156303, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27224249

RESUMO

Sphingosine-1-phosphate receptor 2 (S1PR2) couples with the Gi, Gq, and G12/13 group of proteins, which modulate an array of cellular signaling pathways and affect immune responses to multiple stimuli. In this study, we demonstrated that knockdown of S1PR2 by a specific S1PR2 shRNA lentiviral vector significantly inhibited IL-1ß, IL-6, and TNF-α protein levels induced by oral pathogen Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) in murine bone marrow-derived monocytes and macrophages (BMMs) compared with controls. In addition, knockdown of S1PR2 by the S1PR2 shRNA lentiviral vector suppressed p-PI3K, p-ERK, p-JNK, p-p38, and p-NF-κBp65 protein expressions induced by A. actinomycetemcomitans. Furthermore, bone marrow cells treated with the S1PR2 shRNA lentiviral vector inhibited osteoclastogenesis induced by RANKL compared with controls. The S1PR2 shRNA suppressed the mRNA levels of six osteoclastogenic factors including nuclear factor of activated T-cells cytoplasmic calcineurin-dependent 1 (NFATc1), cathepsin K (Ctsk), acid phosphatase 5 (Acp5), osteoclast-associated receptor (Oscar), dendritic cells specific transmembrane protein (Dcstamp), and osteoclast stimulatory transmembrane protein (Ocstamp) in bone marrow cells. We conclude that S1PR2 plays an essential role in modulating proinflammatory cytokine production and osteoclastogenesis. Blocking S1PR2 signaling might be a novel therapeutic strategy to treat inflammatory bone loss diseases.


Assuntos
Citocinas/metabolismo , Osteoclastos/citologia , Osteogênese , Infecções por Pasteurellaceae/imunologia , Receptores de Lisoesfingolipídeo/genética , Aggregatibacter actinomycetemcomitans/imunologia , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/microbiologia , Osteoclastos/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Esfingosina-1-Fosfato , Fator de Necrose Tumoral alfa/metabolismo
8.
Immunity ; 41(1): 104-15, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25035955

RESUMO

The relationship between dendritic cells (DCs) and macrophages is often debated. Here we ask whether steady-state, lymphoid-tissue-resident conventional DCs (cDCs), plasmacytoid DCs (pDCs), and macrophages share a common macrophage-DC-restricted precursor (MDP). Using new clonal culture assays combined with adoptive transfer, we found that MDP fractions isolated by previous strategies are dominated by precursors of macrophages and monocytes, include some multipotent precursors of other hematopoietic lineages, but contain few precursors of resident cDCs and pDCs and no detectable common precursors restricted to these DC types and macrophages. Overall we find no evidence for a common restricted MDP leading to both macrophages and FL-dependent, resident cDCs and pDCs.


Assuntos
Linhagem da Célula/imunologia , Células Dendríticas/citologia , Tecido Linfoide/citologia , Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/citologia , Transferência Adotiva , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Receptor 1 de Quimiocina CX3C , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Granulócitos/citologia , Granulócitos/imunologia , Fator Estimulador de Colônias de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/imunologia , Monócitos/citologia , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Receptores de Quimiocinas/imunologia
9.
Science ; 344(6186): 921-5, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24812208

RESUMO

Long recognized as an evolutionarily ancient cell type involved in tissue homeostasis and immune defense against pathogens, macrophages are being rediscovered as regulators of several diseases, including cancer. Here we show that in mice, mammary tumor growth induces the accumulation of tumor-associated macrophages (TAMs) that are phenotypically and functionally distinct from mammary tissue macrophages (MTMs). TAMs express the adhesion molecule Vcam1 and proliferate upon their differentiation from inflammatory monocytes, but do not exhibit an "alternatively activated" phenotype. TAM terminal differentiation depends on the transcriptional regulator of Notch signaling, RBPJ; and TAM, but not MTM, depletion restores tumor-infiltrating cytotoxic T cell responses and suppresses tumor growth. These findings reveal the ontogeny of TAMs and a discrete tumor-elicited inflammatory response, which may provide new opportunities for cancer immunotherapy.


Assuntos
Macrófagos/imunologia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/imunologia , Receptores Notch/metabolismo , Transdução de Sinais , Molécula 1 de Adesão de Célula Vascular/metabolismo
10.
J. investig. allergol. clin. immunol ; 24(1): 49-55, ene.-feb. 2014. tab, ilus
Artigo em Inglês | IBECS | ID: ibc-119154

RESUMO

Background and Objective: CD163 is a monocyte/macrophage-specific molecule whose expression is induced by corticosteroids and IL-10. The aim of this study was to evaluate the concentration of soluble CD163 (sCD163) in the induced sputum of asthmatic patients before and after therapy with inhaled corticosteroids (ICSs). Patients and Methods: The study was performed in 24 patients with mild allergic asthma (AAs) and 10 healthy controls (HCs). In 18 AAs, induced sputum and serum samples were obtained before ICS therapy (T0) and 7 days later (T7). In the 6 AAs not treated with ICSs the procedures were performed at T0 and T7. The concentration of sCD163 in sputum and serum samples was evaluated using ELISA. Results: There was no significant difference in mean (SD) baseline serum sCD163 concentration between AAs (1030 [449] ng/mL) and HCs (930 [334.5] ng/mL, P=.530). However, at T0 the mean sputum sCD163 concentration was significantly greater in AAs (4.78 [3.34] ng/mL) than in HCs (1.8 [0.41] ng/mL, P=.009). Treatment with ICSs resulted in a significant increase in sCD163 concentration in sputum (P<.0001) but not in serum (P=.679). No change in sputum or serum sCD163 concentration was detected in AAs who were not treated with ICSs. The change in sputum sCD163 concentration inversely correlated with changes in sputum eosinophilia or exhaled nitric oxide concentration. Conclusions: ICS therapy leads to local upregulation of sCD163 expression, which in turn may participate in the anti-inflammatory effects of ICS therapy (AU)


Antecedentes y Objetivo: El CD163 es una molécula específica de monocitos/macrófagos cuya expresión es inducida por los corticosteroides y la interleucina 10 (IL-10). El objetivo de este estudio fue evaluar la concentración de sCD163 en el esputo inducido de pacientes asmáticos, antes y después del tratamiento con corticosteroides inhalados (ICS). Pacientes y Métodos: El estudio se realizó en 24 pacientes con asma alérgica leve (AA) y 10 controles sanos (HC). En 18 AA, se obtuvieron muestras de esputo y suero antes (T0) y a los 7 días (T7) tras la introducción del tratamiento con ICS. Asimismo, en 6 AA no tratados con ICS se llevaron a cabo los mismos procedimientos en T0 y T7. Se evaluó la concentración de sCD163 en esputo y suero de las muestras mediante ELISA. Resultados: No hubo diferencia en la concentración sérica basal media de sCD163 entre AA (1.030± 449 ng/ml) y los HC (930 ± 334,5 ng/ml, p = 0,530). Sin embargo, en T0 la concentración media de sCD163 esputo fue significativamente mayor en los AA (4,78 ± 3,34 ng/ml) que en HC (1,8 ± 0,41 ng/ml, p = 0,009). El tratamiento con ICS dio lugar a un aumento significativo de la concentración en el esputo de sCD163 (p < 0,0001), pero no en el suero (p = 0,679). No se detectaron diferencias en las concentraciones de sCD163 ni en el suero ni en el esputo del grupo de AA que no fueron tratados con ICS. La concentración sCD163 en esputo se correlacionó inversamente con la eosinofilia en esputo y la concentración NO exhalado. Conclusiones: El tratamiento con ICS induce la expresión local de sCD163, que a su vez puede mediar en el mecanismo anti-inflamatorio de este tratamiento (AU)


Assuntos
Humanos , Corticosteroides/farmacocinética , Escarro/imunologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Asma/imunologia , Administração por Inalação , Hipersensibilidade Respiratória/imunologia
11.
J Biol Chem ; 288(51): 36691-702, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24225954

RESUMO

Tumor-derived exosomes have been shown to induce various immunomodulatory effects. However, the underlying signaling pathways are poorly understood. Here, we analyzed the effects of ex vivo-derived exosomes on monocytic cell differentiation/activation using THP-1 cells as model. We isolated exosomes from various body fluids such as amniotic fluid, liver cirrhosis ascites, and malignant ascites of ovarian cancer patients. We observed that exosomes were internalized by THP-1 cells and induced the production of IL-1ß, TNF-α, and IL-6. Analysis of the signaling pathways revealed a fast triggering of NFκB and a delayed activation of STAT3. Pharmacologic and antibody-blocking experiments showed that the initial production of IL-6 was instrumental for subsequent activation of STAT3. Importantly, triggering of cell signaling was not a unique property of tumor exosomes but was also observed with exosomes of noncancerous origin. Exosomal signaling was TLR-dependent as the knockdown of Toll-like receptor 2 (TLR2) and TLR4 blocked NFκB and STAT3 activation. Similar results were obtained with TLR-neutralizing antibodies. Exosomes also triggered the release of cytokines from mouse bone marrow-derived dendritic cells or macrophages. This process was MyD88-dependent, further supporting a role of TLR signaling. Our results suggest that exosomes trigger TLR-dependent signaling pathways in monocytic precursor cells but possibly also in other immune cells. This process could be important for the induction of immunosuppressive mechanisms during cancer progression and inflammatory diseases.


Assuntos
Citocinas/metabolismo , Exossomos/fisiologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Transdução de Sinais , Receptores Toll-Like/metabolismo , Líquido Amniótico/citologia , Líquido Amniótico/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição STAT3/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
J Immunol ; 191(6): 3192-9, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23935193

RESUMO

Engraftment of human CD34⁺ hematopoietic stem/progenitor cells into immunodeficient mice leads to robust reconstitution of human T and B cells but not monocytes and macrophages. To identify the cause underlying the poor monocyte and macrophage reconstitution, we analyzed human myeloid cell development in humanized mice and found that it was blocked at the promonocyte stage in the bone marrow. Expression of human M-CSF or GM-CSF by hydrodynamic injection of cytokine-encoding plasmid completely abolished the accumulation of promonocytes in the bone marrow. M-CSF promoted the development of mature monocytes and tissue-resident macrophages whereas GM-CSF did not. Moreover, correlating with an increased human macrophages at the sites of infection, M-CSF-treated humanized mice exhibited an enhanced protection against influenza virus and Mycobacterium infection. Our study identifies the precise stage at which human monocyte/macrophage development is blocked in humanized mice and reveals overlapping and distinct functions of M-CSF and GM-CSF in human monocyte and macrophage development. The improved reconstitution and functionality of monocytes/macrophages in the humanized mice following M-CSF expression provide a superior in vivo system to investigate the role of macrophages in physiological and pathological processes.


Assuntos
Diferenciação Celular/imunologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Diferenciação Celular/efeitos dos fármacos , Separação Celular , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Precursoras de Monócitos e Macrófagos/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Nat Immunol ; 14(8): 821-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23812096

RESUMO

Monocytes, macrophages and dendritic cells (DCs) are developmentally related regulators of the immune system that share the monocyte-macrophage DC progenitor (MDP) as a common precursor. Unlike differentiation into DCs, the distal pathways for differentiation into monocytes and monocyte-derived macrophages are not fully elucidated. We have now demonstrated the existence of a clonogenic, monocyte- and macrophage-restricted progenitor cell derived from the MDP. This progenitor was a Ly6C(+) proliferating cell present in the bone marrow and spleen that generated the major monocyte subsets and macrophages, but not DCs or neutrophils. By in-depth quantitative proteomics, we characterized changes in the proteome during monocyte differentiation, which provided insight into the molecular principles of developing monocytes, such as their functional maturation. Thus, we found that monocytes and macrophages were renewed independently of DCs from a committed progenitor.


Assuntos
Medula Óssea/imunologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Proteômica/métodos , Baço/imunologia , Animais , Diferenciação Celular/imunologia , Cromatografia Líquida , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Organismos Livres de Patógenos Específicos , Espectrometria de Massas por Ionização por Electrospray , Baço/citologia , Espectrometria de Massas em Tandem
14.
Immunity ; 38(4): 729-41, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23562161

RESUMO

The therapeutic efficacy of anthracyclines relies on antitumor immune responses elicited by dying cancer cells. How chemotherapy-induced cell death leads to efficient antigen presentation to T cells, however, remains a conundrum. We found that intratumoral CD11c(+)CD11b(+)Ly6C(hi) cells, which displayed some characteristics of inflammatory dendritic cells and included granulomonocytic precursors, were crucial for anthracycline-induced anticancer immune responses. ATP released by dying cancer cells recruited myeloid cells into tumors and stimulated the local differentiation of CD11c(+)CD11b(+)Ly6C(hi) cells. Such cells efficiently engulfed tumor antigens in situ and presented them to T lymphocytes, thus vaccinating mice, upon adoptive transfer, against a challenge with cancer cells. Manipulations preventing tumor infiltration by CD11c(+)CD11b(+)Ly6C(hi) cells, such as the local overexpression of ectonucleotidases, the blockade of purinergic receptors, or the neutralization of CD11b, abolished the immune system-dependent antitumor activity of anthracyclines. Our results identify a subset of tumor-infiltrating leukocytes as therapy-relevant antigen-presenting cells.


Assuntos
Antraciclinas/administração & dosagem , Células Apresentadoras de Antígenos/imunologia , Antineoplásicos/administração & dosagem , Células Dendríticas/imunologia , Neoplasias Experimentais/imunologia , Transferência Adotiva , Animais , Antraciclinas/efeitos adversos , Antígenos Ly/metabolismo , Antígenos de Neoplasias/imunologia , Antineoplásicos/efeitos adversos , Apoptose , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Precursoras de Granulócitos/imunologia , Imunidade Celular , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/imunologia , Neoplasias Experimentais/tratamento farmacológico , Nucleotidases/metabolismo , Receptores Purinérgicos/metabolismo
15.
J Am Heart Assoc ; 2(2): e000065, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23537804

RESUMO

BACKGROUND: Recruitment of macrophage precursors to the adventitia plays a key role in the pathogenesis of abdominal aortic aneurysms (AAAs), but molecular mechanisms remain undefined. The innate immune signaling molecule CD14 was reported to be upregulated in adventitial macrophages in a murine model of AAA and in monocytes cocultured with aortic adventitial fibroblasts (AoAf) in vitro, concurrent with increased interleukin-6 (IL-6) expression. We hypothesized that CD14 plays a crucial role in adventitial macrophage precursor recruitment early during AAA formation. METHODS AND RESULTS: CD14(-/-) mice were resistant to AAA formation induced by 2 different AAA induction models: aortic elastase infusion and systemic angiotensin II (AngII) infusion. CD14 gene deletion led to reduced aortic macrophage infiltration and diminished elastin degradation. Adventitial monocyte binding to AngII-infused aorta in vitro was dependent on CD14, and incubation of human acute monocytic leukemia cell line-1 (THP-1) monocytes with IL-6 or conditioned medium from perivascular adipose tissue (PVAT) upregulated CD14 expression. Conditioned medium from AoAf and PVAT induced CD14-dependent monocyte chemotaxis, which was potentiated by IL-6. CD14 expression in aorta and plasma CD14 levels were increased in AAA patients compared with controls. CONCLUSIONS: These findings link CD14 innate immune signaling via a novel IL-6 amplification loop to adventitial macrophage precursor recruitment in the pathogenesis of AAA.


Assuntos
Aneurisma da Aorta Abdominal/imunologia , Movimento Celular/imunologia , Interleucina-6/imunologia , Receptores de Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Túnica Adventícia/imunologia , Animais , Linhagem Celular Tumoral , Ensaios de Migração de Macrófagos , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imunidade Inata , Macrófagos Peritoneais , Camundongos , Camundongos Transgênicos , Transdução de Sinais/imunologia
16.
J Biol Chem ; 287(31): 25758-69, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22700961

RESUMO

The early initiation phase of acute inflammation is anabolic and primarily requires glycolysis with reduced mitochondrial glucose oxidation for energy, whereas the later adaptation phase is catabolic and primarily requires fatty acid oxidation for energy. We reported previously that switching from the early to the late acute inflammatory response following TLR4 stimulation depends on NAD(+) activation of deacetylase sirtuin 1 (SirT1). Here, we tested whether NAD(+) sensing by sirtuins couples metabolic polarity with the acute inflammatory response. We found in TLR4-stimulated THP-1 promonocytes that SirT1 and SirT 6 support a switch from increased glycolysis to increased fatty acid oxidation as early inflammation converts to late inflammation. Glycolysis enhancement required hypoxia-inducing factor-1α to up-regulate glucose transporter Glut1, phospho-fructose kinase, and pyruvate dehydrogenase kinase 1, which interrupted pyruvate dehydrogenase and reduced mitochondrial glucose oxidation. The shift to late acute inflammation and elevated fatty acid oxidation required peroxisome proliferator-activated receptor γ coactivators PGC-1α and ß to increase external membrane CD36 and fatty acid mitochondrial transporter carnitine palmitoyl transferase 1. Metabolic coupling between early and late responses also required NAD(+) production from nicotinamide phosphoryltransferase (Nampt) and activation of SirT6 to reduce glycolysis and SirT1 to increase fatty oxidation. We confirmed similar shifts in metabolic polarity during the late immunosuppressed stage of human sepsis blood leukocytes and murine sepsis splenocytes. We conclude that NAD(+)-dependent bioenergy shifts link metabolism with the early and late stages of acute inflammation.


Assuntos
Metabolismo Energético , Ácidos Graxos/metabolismo , Glucose/metabolismo , Sepse/metabolismo , Sirtuína 1/metabolismo , Sirtuínas/metabolismo , Adaptação Fisiológica/imunologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Citocinas/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Glicólise , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Células Precursoras de Monócitos e Macrófagos/fisiologia , NAD/biossíntese , Nicotinamida Fosforribosiltransferase/metabolismo , Oxirredução , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas de Ligação a RNA , Sepse/imunologia , Receptor 4 Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
J Innate Immun ; 3(6): 550-64, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22025201

RESUMO

Intestinal macrophages are essential for local homeostasis and in keeping a balance between commensal microbiota and the host. However, they also play essential roles in inflammation and protective immunity, when they change from peaceful regulators to powerful aggressors. As a result, activated macrophages are important targets for treatment of inflammatory bowel diseases such as Crohn's disease. Until recently, the complexity and heterogeneity of intestinal macrophages have been underestimated and here we review current evidence that there are distinct populations of resident and inflammatory macrophages in the intestine. We describe the mechanisms that ensure macrophages remain partially inert in the healthy gut and cannot promote inflammation despite constant exposure to bacteria and other stimuli. This may be because the local environment 'conditions' macrophage precursors to become unresponsive after they arrive in the gut. Nevertheless, this permits some active, physiological functions to persist. A new population of pro-inflammatory macrophages appears in inflammation and we review the evidence that this involves recruitment of a distinct population of fully responsive monocytes, rather than alterations in the existing cells. A constant balance between these resident and inflammatory macrophages is critical for maintaining the status quo in healthy gut and ensuring protective immunity when required.


Assuntos
Infecções Bacterianas/imunologia , Mucosa Intestinal/imunologia , Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/imunologia , Monócitos/imunologia , Animais , Comunicação Celular/imunologia , Movimento Celular/imunologia , Homeostase , Humanos , Tolerância Imunológica , Inflamação , Mucosa Intestinal/microbiologia , Macrófagos/microbiologia , Células Precursoras de Monócitos e Macrófagos/microbiologia , Monócitos/microbiologia
18.
J Hazard Mater ; 196: 335-41, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21974847

RESUMO

Our previous studies found that zinc oxide (ZnO) particles induced expression of intercellular adhesion molecule-1 (ICAM-1) protein in vascular endothelial cells via NF-κB and that zinc ions dissolved from ZnO particles might play the major role in the process. This study aimed to determine if zinc ions could cause inflammatory responses in a human promonocytic leukemia cell line HL-CZ. Conditioned media from the zinc-treated HL-CZ cells induced ICAM-1 protein expression in human umbilical vein endothelial cells (HUVEC). Zinc treatment induced chemokine and inflammatory cytokine release from HL-CZ cells. Inhibition of NFκB activity by over-expression of IκBα in HL-CZ cells did not block the conditioned medium-induced ICAM-1 protein expression in HUVEC cells. Zinc treatment induced activation of multiple immune response-related transcription factors in HL-CZ cells. These results clearly show that zinc ions induce chemokine and inflammatory cytokine release from human promonocytes, accompanied with activation of multiple immune response-related transcription factors. Our in vitro evidence in the zinc-induced inflammatory responses of vascular cells provides a critical linkage between zinc exposure and pathogenesis of those inflammatory vascular diseases.


Assuntos
Quimiocinas/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/imunologia , Zinco/toxicidade , Adenoviridae/genética , Sequência de Bases , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Quimiocinas/genética , Meios de Cultivo Condicionados , Citocinas/genética , Citocinas/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Genes Reporter , Células Endoteliais da Veia Umbilical Humana , Humanos , Luciferases de Vaga-Lume/genética , Dados de Sequência Molecular , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fatores de Transcrição/genética , Acetato de Zinco/toxicidade
19.
Leuk Lymphoma ; 52(1): 92-100, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21219126

RESUMO

Differentiation between acute monoblastic/monocytic leukemia (AMoL) and chronic myelomonocytic leukemia (CMML) can be difficult. Therefore, we compared immunophenotypes between 27 cases of AMoL and 138 cases of CMML. Monocytopoietic cells showed aberrant coexpression of CD56 in all cases of AMoL vs. 81.9% of CMML (p = 0.015). No other aberrantly expressed antigen was found in AMoL, while in CMML CD2 coexpression was found in 21.7% (p = 0.005), lack of CD13 in 10.9%, and of HLA-DR in 4.3% (NS). Cytomorphology identified higher blast percentages and lower percentages of monocytes and granulocytic cells in AMoL (p <0.001). Multiparameter flow-cytometry (MFC) found higher percentages of blasts (17.6 ±â€Š25.2 vs. 4.1 ±â€Š3.2, p <0.001) and monocytopoietic cells (29.1 ±â€Š27.5 vs. 19.9 ±â€Š12.2, p = 0.012) in AMoL and more granulocytic cells in CMML (52.4 ±â€Š19.2 vs. 26.0 ±â€Š22.4, p <0.001). The mean ratio of monocytic:granulocytic cells was higher in AMoL (5.0 vs. 0.8; p <0.001). It can be concluded that AMoL and CMML differ in aberrantly expressed antigens and the amount of granulocytic cells.


Assuntos
Antígenos CD/metabolismo , Granulócitos/metabolismo , Leucemia Monocítica Aguda/metabolismo , Leucemia Monocítica Aguda/patologia , Leucemia Mielomonocítica Crônica/metabolismo , Leucemia Mielomonocítica Crônica/patologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Citometria de Fluxo , Granulócitos/imunologia , Granulócitos/patologia , Humanos , Imunofenotipagem , Leucemia Monocítica Aguda/imunologia , Leucemia Mielomonocítica Crônica/imunologia , Masculino , Pessoa de Meia-Idade , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/patologia , Prognóstico , Adulto Jovem
20.
Cytotherapy ; 12(8): 1022-34, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20849360

RESUMO

BACKGROUND AIMS: Circulating monocytes have been exploited as an important progenitor cell resource for hepatocytes in vitro and are instrumental in the removal of fibrosis. We investigated the significance of monocytes in peripheral blood stem cells (PBSC) for the treatment of liver cirrhosis. METHODS: Rat CD14+ monocytes in PBSC were mobilized with granulocyte-colony-stimulating factor (G-CSF) and harvested by magnetic cell sorting (MACS). Female rats with carbon tetrachloride (CCl4-induced liver cirrhosis were injected CM-DiI-labeled monocytes, CD14⁻ cells (1 x 107 cells/rat) or saline via the portal vein. RESULTS: Rat CD14+ and CD11b+ monocytes in PBSC were partly positive for CD34, CD45, CD44, Oct3/4 and Sox2, suggesting monocytes with progenitor capacity. Compared with CD14⁻ cell-infused and saline-injected rats, rats undergoing monocyte transplantation showed a gradually increased serum albumin level and decreased portal vein pressure, resulting in a significantly improved survival rate. Meanwhile, monocyte transplantation apparently attenuated liver fibrosis by analysis for fibronectin, α2-(1)-procollagen, α-smooth muscle aorta (SMA) and transforming growth factor (TGF)-ß. Transplanted monocytes mainly clustered in periportal areas of liver, in which 1.8% cells expressed hepatocyte marker albumin and CK18. The expression level of hepatocyte growth factor (HGF), TGF-α, extracellular matrix (EGF) and vascular endothelial growth factor (VEGF) increased, while monocyte transplantation enhanced hepatocyte proliferation. On the other hand, the activities and expression of matrix metalloproteinases (MMP) increased while tissue inhibitor of metalloproteinase (TIMP)-1 expression significantly reduced in monocyte-transplanted livers. Some transplanted monocytes expressed MMP-9 and -13. CONCLUSIONS: The data suggest that CD14+ monocytes in PBSC contribute to hepatocyte regeneration and extracellular matrix (ECM) remodeling in rat liver cirrhosis much more than CD14⁻ cells, and might offer a therapeutic alternative for patients with liver cirrhosis.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Hepatócitos/metabolismo , Cirrose Hepática/terapia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Animais , Biomarcadores/metabolismo , Tetracloreto de Carbono/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Receptores de Lipopolissacarídeos/biossíntese , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Cirrose Hepática/fisiopatologia , Cirrose Hepática/prevenção & controle , Masculino , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/imunologia , Células Precursoras de Monócitos e Macrófagos/patologia , Ratos , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...