Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Sci Rep ; 10(1): 11933, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32686739

RESUMO

Loss of estrogens at menopause is a major cause of osteoporosis and increased fracture risk. Estrogens protect against bone loss by decreasing osteoclast number through direct actions on cells of the myeloid lineage. Here, we investigated the molecular mechanism of this effect. We report that 17ß-estradiol (E2) decreased osteoclast number by promoting the apoptosis of early osteoclast progenitors, but not mature osteoclasts. This effect was abrogated in cells lacking Bak/Bax-two pro-apoptotic members of the Bcl-2 family of proteins required for mitochondrial apoptotic death. FasL has been previously implicated in the pro-apoptotic actions of E2. However, we show herein that FasL-deficient mice lose bone mass following ovariectomy indistinguishably from FasL-intact controls, indicating that FasL is not a major contributor to the anti-osteoclastogenic actions of estrogens. Instead, using microarray analysis we have elucidated that ERα-mediated estrogen signaling in osteoclast progenitors decreases "oxidative phosphorylation" and the expression of mitochondria complex I genes. Additionally, E2 decreased the activity of complex I and oxygen consumption rate. Similar to E2, the complex I inhibitor Rotenone decreased osteoclastogenesis by promoting osteoclast progenitor apoptosis via Bak/Bax. These findings demonstrate that estrogens decrease osteoclast number by attenuating respiration, and thereby, promoting mitochondrial apoptotic death of early osteoclast progenitors.


Assuntos
Trifosfato de Adenosina/biossíntese , Estrogênios/metabolismo , Mitocôndrias/metabolismo , Células Precursoras de Monócitos e Macrófagos/metabolismo , Osteoclastos/metabolismo , Fosforilação Oxidativa , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Densidade Óssea , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Contagem de Células , Diferenciação Celular , Células Cultivadas , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais
2.
Exp Cell Res ; 390(1): 111932, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32145253

RESUMO

Patients with estrogen receptor α positive (ERα+) breast cancer can respond to endocrine therapy, but treatment resistance is common and associated with downregulation of ERα expression in the dormant residual cells. Here we show, using long-term NSG xenograft models of human breast cancer and primary human monocytes, in vitro primary cell cultures and tumors from breast cancer patients, that macrophage derived tumor necrosis factor alpha (TNFα) downregulates ERα in breast cancer cells via inactivation of the transcription factor Forkhead box O transcription factor 3a (FOXO3a). Moreover, presence of tumor associated macrophages in the primary tumor of breast cancer patients, was associated with ERα negativity, and with worse prognosis in patients with ERα+ tumors. We propose that pro-inflammatory macrophages, despite being tumoricidal, may have direct effects on tumor progression and endocrine resistance in breast cancer patients. Our findings suggest that TNFα antagonists should be evaluated for treatment of ERα+ breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/genética , Proteína Forkhead Box O3/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Neoplasias da Mama/genética , Células Cultivadas , Regulação para Baixo , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Macrófagos/citologia , Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Células Precursoras de Monócitos e Macrófagos/transplante
3.
FASEB J ; 33(10): 11006-11020, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31284764

RESUMO

Monocytes/macrophages drive progression and regression of atherosclerosis. Conjugated linoleic acid (CLA), an anti-inflammatory lipid, mediates atheroprotective effects. We investigated how CLA alters monocyte/macrophage phenotype during attenuated progression and regression of atherosclerosis. Apolipoprotein E knockout (ApoE-/-) mice were fed a high-fat (60%) high-cholesterol (1%) diet (HFHCD) for 2 wk, followed by 6-wk 1% CLA 80:20 supplementation to investigate disease progression. Simultaneously, ApoE-/- mice were fed a 12-wk HFHCD with/without CLA for the final 4 wk to investigate regression. Aortic lesions were quantified by en face staining. Proteomic analysis, real-time quantitative PCR and flow cytometry were used to interrogate monocyte/macrophage phenotypes. CLA supplementation inhibited atherosclerosis progression coincident with decreased proinflammatory and increased anti-inflammatory macrophages. However, CLA-induced regression was associated with increased proinflammatory monocytes resulting in increased proresolving M2 bone marrow-derived macrophages, splenic macrophages, and dendritic cells in lesion-draining lymph nodes. Proteomic analysis confirmed regulation of a proinflammatory bone marrow response, which was abolished upon macrophage differentiation. Thus, in attenuation and regression of atherosclerosis, regardless of the monocyte signature, during monocyte to macrophage differentiation, proresolving macrophages prevail, mediating vascular repair. This study provides novel mechanistic insight into the monocyte/macrophage phenotypes in halted atherosclerosis progression and regression of atherosclerosis.-Bruen, R., Curley, S., Kajani, S., Lynch, G., O'Reilly, M. E., Dillon, E. T., Fitzsimons, S., Mthunzi, L., McGillicuddy, F. C., Belton, O. Different monocyte phenotypes result in proresolving macrophages in conjugated linoleic acid-induced attenuated progression and regression of atherosclerosis.


Assuntos
Aterosclerose/tratamento farmacológico , Diferenciação Celular , Ácidos Linoleicos Conjugados/farmacologia , Fenótipo , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/metabolismo , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Ácidos Linoleicos Conjugados/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/metabolismo , Proteoma/genética , Proteoma/metabolismo
4.
FASEB J ; 33(10): 11082-11095, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31295022

RESUMO

JNK1 plays an important role in osteoclastogenesis in response to the osteoclastogenic cytokine receptor activator for nuclear factor-κB ligand (RANKL). JNK1 is widely accepted as an autophagy regulator under stress conditions. However, the role of JNK1-mediated autophagy in osteoclastogenesis remains largely unknown. In the current study, our data showed that JNK1 inhibition by a pharmacological inhibitor or RNA interference significantly reduced the autophagic response induced by RANKL in osteoclast precursors (OCPs) derived from bone marrow-derived macrophages. Overexpression of the key autophagy protein Beclin1 rescued autophagy deficiency and osteoclastogenesis in the presence of a JNK inhibitor (SP600125). In contrast, JNK activator (anisomycin)-induced autophagy was blocked by Beclin1 knockdown in OCPs. In addition, JNK1 inhibition increased apoptosis and blocked autophagy, whereas overexpression of Beclin1 reversed the enhanced apoptosis induced by JNK1 inhibition in OCPs. Furthermore, RANKL could induce the phosphorylation of Bcl-2, subsequently dissociating Beclin1 from the Bcl-2-Beclin1 complex, which could be blocked by JNK1 inhibition. Collectively, this study revealed that JNK1 regulated osteoclastogenesis by activating Bcl-2-Beclin1-autophagy signaling in addition to the classic c-Jun/activator protein 1 pathway, which provided the first evidence for the contribution of JNK1 signaling to OCP autophagy and the autophagic mechanism underlying JNK1-regulated osteoclastogenesis. An important osteoclastogenesis-regulating signaling pathway (JNK1-Bcl-2-Beclin1-autophagy activation) was identified, which provides novel potential targets for the clinical therapy of metabolic bone diseases.-Ke, D., Ji, L., Wang, Y., Fu, X., Chen, J., Wang, F., Zhao, D., Xue, Y., Lan, X., Hou, J. JNK1 regulates RANKL-induced osteoclastogenesis via activation of a novel Bcl-2-Beclin1-autophagy pathway.


Assuntos
Autofagia , Diferenciação Celular , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Animais , Apoptose , Proteína Beclina-1/metabolismo , Células Cultivadas , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Osteoblastos/citologia , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células RAW 264.7 , Transdução de Sinais
5.
FEBS J ; 286(17): 3433-3449, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31044513

RESUMO

Hyaluronan is a glycosaminoglycan normally present in the extracellular matrix in most tissues. Hyaluronan is a crucial player in many processes associated with cancer, such as angiogenesis, invasion, and metastasis. However, little has been reported regarding the action of hyaluronan on monocytes/macrophages (Mo/MØ) in tumor angiogenesis and its consequences on tumor development. In the present study, we investigated the effects of hyaluronan of different sizes on human Mo/MØ angiogenic behavior in colorectal and breast carcinoma. In vitro, the treatment of Mo/MØ with lysates and conditioned media from a breast but not from colorectal carcinoma cell line plus high-molecular weight hyaluronan induced: (a) an increased expression of angiogenic factors VEGF, IL-8, FGF-2, and MMP-2, (b) an increased endothelial cell migration, and (c) a differential expression of hyaluronan-binding protein TSG-6. Similar results were observed in Mo/MØ derived from breast cancer patients treated with tumor lysates. Besides, macrophages primed with high-molecular weight hyaluronan and inoculated in human breast cancer xenograft tumor increased blood vessel formation and diminished TSG-6 levels. In contrast, the effects triggered by high-molecular weight hyaluronan on Mo/MØ in breast cancer context were not observed in the context of colorectal carcinoma. Taken together, these results indicate that the effect of high-molecular weight hyaluronan as an inductor of the angiogenic behavior of macrophages in breast tumor context is in part consequence of the presence of TSG-6.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Moléculas de Adesão Celular/genética , Neoplasias Colorretais/metabolismo , Ácido Hialurônico/farmacologia , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Neovascularização Patológica/metabolismo , Animais , Neoplasias da Mama/patologia , Carcinoma/patologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Meios de Cultivo Condicionados/farmacologia , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Curr Opin Immunol ; 50: 64-74, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29247852

RESUMO

The combination between novel fate-mapping tools and single-cell RNA-sequencing technology has revealed the presence of multiple macrophage progenitors. This raises the fascinating possibility that what was once perceived as immense functional plasticity of macrophages could in fact come down to separate macrophage subsets performing distinct functions because of their differential cellular origin. The question of macrophage plasticity versus macrophage heterogeneity is broader than the difference between macrophages of embryonic or adult hematopoietic origin and is particularly relevant in the context of inflammation. In this manuscript, we review the potential impact of cellular origin on the function of macrophages. We also highlight the need for novel 'functional fate-mapping' tools that would reveal the history of the functional state of macrophages, rather than their cellular origin, in order to finally study their true plasticity in vivo.


Assuntos
Diferenciação Celular , Plasticidade Celular , Macrófagos/citologia , Macrófagos/fisiologia , Animais , Biomarcadores , Diferenciação Celular/imunologia , Plasticidade Celular/imunologia , Suscetibilidade a Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fenótipo
7.
Proc Natl Acad Sci U S A ; 114(50): 13296-13301, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29167373

RESUMO

Epitranscriptomics refers to posttranscriptional alterations on an mRNA sequence that are dynamic and reproducible, and affect gene expression in a similar way to epigenetic modifications. However, the functional relevance of those modifications for the transcript, the cell, and the organism remain poorly understood. Here, we focus on RNA editing and show that Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-1 (APOBEC1), together with its cofactor RBM47, mediates robust editing in different tissues. The majority of editing events alter the sequence of the 3'UTR of targeted transcripts, and we focus on one cell type (monocytes) and on a small set of highly edited transcripts within it to show that editing alters gene expression by modulating translation (but not RNA stability or localization). We further show that specific cellular processes (phagocytosis and transendothelial migration) are enriched for transcripts that are targets of editing and that editing alters their function. Finally, we survey bone marrow progenitors and demonstrate that common monocyte progenitor cells express high levels of APOBEC1 and are susceptible to loss of the editing enzyme. Overall, APOBEC1-mediated transcriptome diversification is required for the fine-tuning of protein expression in monocytes, suggesting an epitranscriptomic mechanism for the proper maintenance of homeostasis in innate immune cells.


Assuntos
Desaminase APOBEC-1/metabolismo , Epigênese Genética , Edição de RNA , Transcriptoma , Desaminase APOBEC-1/genética , Animais , Movimento Celular , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fagocitose
8.
Immunity ; 46(5): 835-848.e4, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28514689

RESUMO

Monocytes give rise to macrophages and dendritic cells (DCs) under steady-state and inflammatory conditions, thereby contributing to host defense and tissue pathology. A common monocyte progenitor (cMoP) that is strictly committed to the monocyte lineage has been recently identified in mice. Here, we identified human cMoPs as a CLEC12AhiCD64hi subpopulation of conventional granulocyte-monocyte progenitors (cGMPs) in umbilical cord blood and in bone marrow. Human cMoPs gave rise to monocyte subsets without showing any potential for differentiating into myeloid or lymphoid cells. Within the cGMP population, we also identified revised GMPs that completely lacked DC and lymphoid potential. Collectively, our findings expand and revise the current understanding of human myeloid cell differentiation pathways.


Assuntos
Diferenciação Celular , Evolução Clonal , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Animais , Antígenos CD/metabolismo , Biomarcadores , Ciclo Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Análise por Conglomerados , Citocinas/metabolismo , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Camundongos
9.
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
10.
Methods Mol Biol ; 1371: 89-100, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26530796

RESUMO

In the last years, cell therapy has become a promising approach to therapeutically manipulate immune responses in autoimmunity, cancer, and transplantation. Several types of lymphoid and myeloid cells origin have been generated in vitro and tested in animal models. Their efficacy to decrease pharmacological treatment has successfully been established. Macrophages play an important role in physiological and pathological processes. They represent an interesting cell population due to their high plasticity in vivo and in vitro. Here, we describe a protocol to differentiate murine regulatory macrophages in vitro from bone marrow precursors. We also describe several methods to assess macrophage classical functions, as their bacterial killing capacity and antigen endocytosis and degradation. Importantly, regulatory macrophages also display suppressive characteristics, which are addressed by the study of their hypostimulatory T lymphocyte capacity and polyclonal T lymphocyte activation suppression.


Assuntos
Células da Medula Óssea/citologia , Imunomodulação , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Diferenciação Celular , Técnicas de Cocultura , Citotoxicidade Imunológica , Endocitose/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Células Precursoras de Monócitos e Macrófagos/citologia
11.
Immunity ; 41(1): 5-7, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25035946

RESUMO

Macrophage and dendritic cell (DC) progenitors (MDPs) produce macrophages and DCs but not other hematopoietic lineages. In this issue of Immunity, Sathe et al. (2014) show that isolated MDP populations hardly contain such bipotent progenitors at clonal levels, arguing against the existence of MDPs.


Assuntos
Linhagem da Célula/imunologia , Células Dendríticas/citologia , Tecido Linfoide/citologia , Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/citologia , Animais
12.
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
13.
Am J Physiol Heart Circ Physiol ; 307(5): H762-72, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24993041

RESUMO

The main objective of this study was to determine whether or not monocyte infiltration occurs in the prediabetic (PD) heart and its role in PD cardiomyopathy. We hypothesized that the PD heart is significantly populated with monocytes and that bone morphogenetic protein (BMP)-7, a novel mediator of monocyte polarization, activates infiltrated monocytes into anti-inflammatory M2 macrophages, thereby inhibiting apoptosis and fibrosis and improving cardiac function. C57Bl6 mice were assigned to control, PD, or PD + BMP-7 groups. PD and PD + BMP-7 groups were administered streptozotocin (50 mg/kg), whereas control animals received sodium citrate buffer. Afterward, the PD + BMP-7 group was administered BMP-7 (200 µg/kg) for 3 days. Our data showed significantly increased infiltrated monocytes and associated pro-inflammatory cytokines, adverse cardiac remodeling, and heart dysfunction in the PD group (P < 0.05). Interestingly, M2 macrophage differentiation and associated anti-inflammatory cytokines were enhanced and there were reduced adverse cardiac remodeling and improved cardiac function in the PD + BMP-7 group (P < 0.05). In conclusion, our data suggest that PD cardiomyopathy is associated with increased monocyte infiltration and released proinflammatory cytokines, which contributes to adverse cardiac remodeling and cardiac dysfunction. Moreover, we report that BMP-7 possesses novel therapeutic potential in its ability to differentiate monocytes into M2 macrophages and confer cardiac protection in the PD heart.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Cardiomiopatias Diabéticas/tratamento farmacológico , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Apoptose , Proteína Morfogenética Óssea 7/uso terapêutico , Diferenciação Celular , Movimento Celular , Cardiomiopatias Diabéticas/patologia , Fibrose/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/fisiologia
14.
Semin Immunopathol ; 36(2): 137-48, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24435095

RESUMO

Monocytes are mononuclear circulating phagocytes that originate in the bone marrow and give rise to macrophages in peripheral tissue. For decades, our understanding of monocyte lineage was bound to a stepwise model that favored an inverse relationship between cellular proliferation and differentiation. Sophisticated molecular and surgical cell tracking tools have transformed our thinking about monocyte topo-ontogeny and function. Here, we discuss how recent studies focusing on progenitor proliferation and differentiation, monocyte mobilization and recruitment, and macrophage differentiation and proliferation are reshaping knowledge of monocyte lineage in steady state and disease.


Assuntos
Monócitos/imunologia , Monócitos/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Quimiotaxia de Leucócito/imunologia , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/metabolismo , Monócitos/citologia , Fagócitos/citologia , Fagócitos/imunologia , Fagócitos/metabolismo
15.
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
16.
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
17.
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
18.
Cell Biochem Biophys ; 67(3): 997-1004, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575939

RESUMO

We sought to investigate whether TSG suppressed the ICAM-1/VCAM-1 expression in dietary atherosclerotic rats and in Ox-LDL-induced U937 cells. For this purpose, 60 male Sprague-Dawley rats were randomly-and-equally divided into six groups. Atherosclerosis was induced by feeding rats a hyperlipidemic diet. TSG (120, 60 or 30 mg/kg/day) was administered by oral gavage. Simvastatin (2 mg/kg/day) was administered as positive control whereas physiological saline (0.9% NaCl) served as untreated control. After 12 weeks, rats were euthanized by ethyl carbonate (1,200 mg/kg) and aortic wall samples were collected. Besides, U937 cells were stimulated for 48 h by Ox-LDL (80 µg/mL) with and without TSG (120, 60, 30 µg/L) or simvastatin (100 µg/L). ICAM-1/VCAM-1 mRNA expression was determined by RT-PCR and protein expression was detected by immunohistochemistry and/or western blotting. The data show that ICAM-1/VCAM-1 mRNA/protein expression was significantly enhanced in atherosclerotic aortas compared with normal diet group. Ox-LDL-induced ICAM-1/VCAM-1 mRNA/protein expression in U937 cells. Importantly, TSG significantly inhibited ICAM-1/VCAM-1 expression in atherosclerotic aortas in a dose-dependent manner. TSG-pretreatment also inhibited ICAM-1/VCAM-1 expression in Ox-LDL-induced U937 cells. Therefore, we concluded that TSG suppressed the expression of adhesion (ICAM-1/VCAM-1) molecules both in vivo (in aortic wall of dietary atherosclerotic rats) and in vitro (U937 cells).


Assuntos
Aorta/efeitos dos fármacos , Aterosclerose/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucosídeos/farmacologia , Molécula 1 de Adesão Intercelular/metabolismo , Estilbenos/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Animais , Aorta/metabolismo , Aterosclerose/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Dieta Hiperlipídica , Humanos , Lipoproteínas LDL/toxicidade , Masculino , Células Precursoras de Monócitos e Macrófagos/citologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sinvastatina/farmacologia , Células U937
19.
PLoS One ; 7(10): e47930, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110133

RESUMO

Osteoclasts are unique multinucleated cells formed by fusion of preosteoclasts derived from cells of the monocyte/macrophage lineage, which are induced by RANKL. However, characteristics and subpopulations of osteoclast precursor cells are poorly understood. We show here that a combination of TNF-α, TGF-ß, and M-CSF efficiently generates mononuclear preosteoclasts but not multinucleated osteoclasts (MNCs) in rat bone marrow cultures depleted of stromal cells. Using a rat osteoclast-specific mAb, Kat1, we found that TNF-α and TGF-ß specifically increased Kat1(+)c-fms(+) and Kat1(+)c-fms(-) cells but not Kat1(-)c-fms(+) cells. Kat1(-)c-fms(+) cells appeared in early stages of culture, but Kat1(+)c-fms(+) and Kat1(+)c-fms(-) cells increased later. Preosteoclasts induced by TNF-α, TGF-ß, and M-CSF rapidly differentiated into osteoclasts in the presence of RANKL and hydroxyurea, an inhibitor of DNA synthesis, suggesting that preosteoclasts are terminally differentiated cells. We further analyzed the expression levels of genes encoding surface proteins in bone marrow macrophages (BMM), preosteoclasts, and MNCs. Preosteoclasts expressed itgam (CD11b) and chemokine receptors CCR1 and CCR2; however, in preosteoclasts the expression of chemokine receptors CCR1 and CCR2 was not up-regulated compared to their expression in BMM. However, addition of RANKL to preosteoclasts markedly increased the expression of CCR1. In contrast, expression of macrophage antigen emr-1 (F4/80) and chemokine receptor CCR5 was down-regulated in preosteoclasts. The combination of TNF-α, TGF-ß, and M-CSF induced Kat1(+)CD11b(+) cells, but these cells were also induced by TNF-α alone. In addition, MIP-1α and MCP-1, which are ligands for CCR1 and CCR2, were chemotactic for preosteoclasts, and promoted multinucleation of preosteoclasts. Finally, we found that Kat1(+)c-fms(+) cells were present in bone tissues of rats with adjuvant arthritis. These data demonstrate that TNF-α in combination with TGF-ß efficiently generates preosteoclasts in vitro. We delineated characteristics that are useful for identifying and isolating rat preosteoclasts, and found that CCR1 expression was regulated in the fusion step in osteoclastogenesis.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/citologia , Osteoclastos/citologia , Animais , Anticorpos Monoclonais , Diferenciação Celular/fisiologia , Primers do DNA/genética , Citometria de Fluxo , Imunofluorescência , Hidroxiureia/farmacologia , Imuno-Histoquímica , Fator Estimulador de Colônias de Macrófagos/farmacologia , Ligante RANK/farmacologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptores CCR1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
20.
Bull Exp Biol Med ; 152(4): 489-93, 2012 Feb.
Artigo em Inglês, Russo | MEDLINE | ID: mdl-22803118

RESUMO

The effects of pulmonary surfactant on the morphology and functioning of young macrophages were studied on the model of monocyte/macrophage differentiation in vitro and on macrophages of the bronchial alveolar lavage fluid. Surfactant is not a differentiation inductor, but it stimulated the maturation and phagocytic activity of young macrophages. The stimulatory effect of surfactant on phagocytic activity of macrophages persisted even after its removal from the culture medium.


Assuntos
Macrófagos Alveolares/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Surfactantes Pulmonares/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Macrófagos Alveolares/citologia , Células Precursoras de Monócitos e Macrófagos/citologia , Monócitos/citologia , Fagocitose/efeitos dos fármacos , Ésteres de Forbol , Tuberculose Pulmonar/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...