Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Immunol ; 12: 618081, 2021.
Article in English | MEDLINE | ID: mdl-33692791

ABSTRACT

As hematopoietic progenitors supply a large number of blood cells, therapeutic strategies targeting hematopoietic progenitors are potentially beneficial to eliminate unwanted blood cells, such as leukemic cells and immune cells causing diseases. However, due to their pluripotency, targeting those cells may impair the production of multiple cell lineages, leading to serious side effects such as anemia and increased susceptibility to infection. To minimize those side effects, it is important to identify monopotent progenitors that give rise to a particular cell lineage. Monocytes and monocyte-derived macrophages play important roles in the development of inflammatory diseases and tumors. Recently, we identified human monocyte-restricted progenitors, namely, common monocyte progenitors and pre-monocytes, both of which express high levels of CD64, a well-known monocyte marker. Here, we introduce a dimeric pyrrolobenzodiazepine (dPBD)-conjugated anti-CD64 antibody (anti-CD64-dPBD) that selectively induces the apoptosis of proliferating human monocyte-restricted progenitors but not non-proliferating mature monocytes. Treatment with anti-CD64-dPBD did not affect other types of hematopoietic cells including hematopoietic stem and progenitor cells, neutrophils, lymphocytes and platelets, suggesting that its off-target effects are negligible. In line with these findings, treatment with anti-CD64-dPBD directly killed proliferating monocytic leukemia cells and prevented monocytic leukemia cell generation from bone marrow progenitors of chronic myelomonocytic leukemia patients in a patient-derived xenograft model. Furthermore, by depleting the source of monocytes, treatment with anti-CD64-dPBD ultimately eliminated tumor-associated macrophages and significantly reduced tumor size in humanized mice bearing solid tumors. Given the selective action of anti-CD64-dPBD on proliferating monocyte progenitors and monocytic leukemia cells, it should be a promising tool to target cancers and other monocyte-related inflammatory disorders with minimal side effects on other cell lineages.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/pharmacology , Monocyte-Macrophage Precursor Cells/drug effects , Animals , Antineoplastic Agents, Immunological/therapeutic use , Humans , Immunoconjugates/therapeutic use , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , Monocyte-Macrophage Precursor Cells/metabolism , Monocytes/drug effects , Monocytes/metabolism , THP-1 Cells , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism
2.
Sci Rep ; 10(1): 11933, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32686739

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate/biosynthesis , Estrogens/metabolism , Mitochondria/metabolism , Monocyte-Macrophage Precursor Cells/metabolism , Osteoclasts/metabolism , Oxidative Phosphorylation , Animals , Apoptosis/drug effects , Biomarkers , Bone Density , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Count , Cell Differentiation , Cells, Cultured , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Mice , Mice, Knockout , Mitochondria/drug effects , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/drug effects , Signal Transduction
3.
Int J Lab Hematol ; 42(4): 418-422, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32297416

ABSTRACT

Enumeration of blasts and promonocytes is essential for World Health Organization (WHO) classification of myelomonocytic neoplasms. The accuracy of distinguishing blasts, promonocytes and monocytes, including normal vs abnormal monocytes, remains controversial. The objective of this analysis is to assess concordances between experienced hematopathologists in classifying cells as blasts, promonocytes, and monocytes according to WHO criteria. Each of 11 hematopathologists assessed glass slides from 20 patients [12 with chronic myelomonocytic leukemia (CMML) and 8 with acute myeloid leukemia (AML)] including blood and BM aspirate smears, and limited nonspecific esterase (NSE) stains. All cases were blindly reviewed. Fleiss' extension of Cohen's kappa for multiple raters was used on these variables, separately for peripheral blood (PB) and bone marrow (BM). Spearman's rank correlation was used to assess correlations between each pair of hematopathologists for each measurement. For the classification based on the sum of blasts and promonocytes in the BM, Fleiss' kappa was estimated as 0.744. For PB, categorizing patients according to the sum of blasts and promonocytes, Fleiss' kappa was estimated as 0.949. Distinction of abnormal monocytes from normal monocytes in PB did not achieve a good concordance and showed strong evidence of differences between hematopathologists (P < .0001). The hematopathologists achieved a good concordance rate of 74% in CMML vs AML classification and a high k rate, confirming that criteria for defining the blasts equivalents (blasts plus promonocytes) could be applied consistently. Identification of monocyte subtypes (abnormal vs normal) was not concordant. Our results support the practice of combining blasts/promonocytes into a single category.


Subject(s)
Blast Crisis , Bone Marrow , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myelomonocytic, Chronic , Monocyte-Macrophage Precursor Cells , Adult , Blast Crisis/classification , Blast Crisis/metabolism , Blast Crisis/pathology , Bone Marrow/metabolism , Bone Marrow/pathology , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/classification , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelomonocytic, Chronic/classification , Leukemia, Myelomonocytic, Chronic/metabolism , Leukemia, Myelomonocytic, Chronic/pathology , Male , Middle Aged , Monocyte-Macrophage Precursor Cells/classification , Monocyte-Macrophage Precursor Cells/metabolism , Monocyte-Macrophage Precursor Cells/pathology
4.
Exp Cell Res ; 390(1): 111932, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32145253

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/genetics , Forkhead Box Protein O3/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Breast Neoplasms/genetics , Cells, Cultured , Down-Regulation , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Macrophages/cytology , Macrophages/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Monocyte-Macrophage Precursor Cells/transplantation
5.
FASEB J ; 33(10): 11006-11020, 2019 10.
Article in English | MEDLINE | ID: mdl-31284764

ABSTRACT

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.


Subject(s)
Atherosclerosis/drug therapy , Cell Differentiation , Linoleic Acids, Conjugated/pharmacology , Phenotype , Animals , Aorta/drug effects , Aorta/metabolism , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Linoleic Acids, Conjugated/therapeutic use , Male , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/metabolism , Proteome/genetics , Proteome/metabolism
6.
FASEB J ; 33(10): 11082-11095, 2019 10.
Article in English | MEDLINE | ID: mdl-31295022

ABSTRACT

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.


Subject(s)
Autophagy , Cell Differentiation , Mitogen-Activated Protein Kinase 8/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , Animals , Apoptosis , Beclin-1/metabolism , Cells, Cultured , Mice , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , RAW 264.7 Cells , Signal Transduction
7.
FEBS J ; 286(17): 3433-3449, 2019 09.
Article in English | MEDLINE | ID: mdl-31044513

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Adhesion Molecules/genetics , Colorectal Neoplasms/metabolism , Hyaluronic Acid/pharmacology , Monocyte-Macrophage Precursor Cells/drug effects , Neovascularization, Pathologic/metabolism , Animals , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Mol Metab ; 20: 38-50, 2019 02.
Article in English | MEDLINE | ID: mdl-30553769

ABSTRACT

OBJECTIVE: Sympathetic nervous system and immune cell interactions play key roles in the regulation of metabolism. For example, recent convergent studies have shown that macrophages regulate obesity through brown adipose tissue (BAT) activation and beiging of white adipose tissue (WAT) via effects upon local catecholamine availability. However, these studies have raised issues about the underlying mechanisms involved including questions regarding the production of catecholamines by macrophages, the role of macrophage polarization state and the underlying intracellular signaling pathways in macrophages that might mediate these effects. METHODS: To address such issues we generated mice lacking Irs2, which mediates the effects of insulin and interleukin 4, specifically in LyzM expressing cells (Irs2LyzM-/- mice). RESULTS: These animals displayed obesity resistance and preservation of glucose homeostasis on high fat diet feeding due to increased energy expenditure via enhanced BAT activity and WAT beiging. Macrophages per se did not produce catecholamines but Irs2LyzM-/- mice displayed increased sympathetic nerve density and catecholamine availability in adipose tissue. Irs2-deficient macrophages displayed an anti-inflammatory transcriptional profile and alterations in genes involved in scavenging catecholamines and supporting increased sympathetic innervation. CONCLUSIONS: Our studies identify a critical macrophage signaling pathway involved in the regulation of adipose tissue sympathetic nerve function that, in turn, mediates key neuroimmune effects upon systemic metabolism. The insights gained may open therapeutic opportunities for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Insulin Receptor Substrate Proteins/genetics , Monocyte-Macrophage Precursor Cells/metabolism , Obesity/genetics , Sympathetic Nervous System/metabolism , Adipose Tissue, Brown/physiology , Animals , Catecholamines/metabolism , Cells, Cultured , Energy Metabolism , Gene Deletion , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Sympathetic Nervous System/physiology
9.
Redox Biol ; 17: 400-410, 2018 07.
Article in English | MEDLINE | ID: mdl-29879549

ABSTRACT

Autophagy has been shown to be stimulated in advanced atherosclerotic plaques by metabolic stress, inflammation and oxidized lipids. The lack of published studies addressing the potential stimulation of pro-survival autophagy by oxysterols, a family of cholesterol oxidation products, has prompted our study. Thus, the goal of the current study is to elucidate the molecular mechanism of the autophagy induced by 27-hydroxycholesterol (27-OH), that is one of the most abundant oxysterols in advanced atherosclerotic lesions, and to assess whether the pro-oxidant effect of the oxysterol is involved in the given response. Here we showed that 27-OH, in a low micromolar range, activates a pro-survival autophagic response in terms of increased LC3 II/LC3 I ratio and Beclin 1, that depends on the up-regulation of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt pathways as a potential result of an intracellular reactive oxygen species increase provoked by the oxysterol in human promonocytic U937 cells. Moreover, 27-OH induced autophagy is dependent on the relation between nuclear factor erythroid 2 p45-related factor 2 (Nrf2)-dependent antioxidant response and p62. The data obtained highlight the involvement of cholesterol oxidation products in the pathogenesis of oxidative stress related chronic diseases like atherosclerosis. Therefore, deeply understanding the complex mechanism and generating synthetic or natural molecules targeting this survival mechanism might be very promising tools in the prevention of such diseases.


Subject(s)
Autophagy/drug effects , Cholesterol/metabolism , Hydroxycholesterols/pharmacology , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/genetics , Antioxidants/pharmacology , Apoptosis/drug effects , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Autophagy/genetics , Cell Survival/drug effects , Humans , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism
10.
Bioinformatics ; 34(7): 1174-1182, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29186334

ABSTRACT

Motivation: Blood cell formation has been recognized as a suitable system to study celular differentiation mainly because of its experimental accessibility, and because it shows characteristics such as hierarchical and gradual bifurcated patterns of commitment, which are present in several developmental processes. Although hematopoiesis has been extensively studied and there is a wealth of molecular and cellular data about it, it is not clear how the underlying molecular regulatory networks define or restrict cellular differentiation processes. Here, we infer the molecular regulatory network that controls the differentiation of a blood cell subpopulation derived from the granulocyte-monocyte precursor (GMP), comprising monocytes, neutrophils, eosinophils, basophils and mast cells. Results: We integrate published qualitative experimental data into a model to describe temporal expression patterns observed in GMP-derived cells. The model is implemented as a Boolean network, and its dynamical behavior is studied. Steady states of the network can be clearly identified with the expression profiles of monocytes, mast cells, neutrophils, basophils, and eosinophils, under wild-type and mutant backgrounds. Availability and implementation: All scripts are publicly available at https://github.com/caramirezal/RegulatoryNetworkGMPModel. Contact: lmendoza@biomedicas.unam.mx. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Cell Differentiation , Computational Biology/methods , Gene Regulatory Networks , Models, Biological , Monocyte-Macrophage Precursor Cells/physiology , Animals , Hematopoiesis , Humans , Monocyte-Macrophage Precursor Cells/metabolism
11.
Curr Opin Immunol ; 50: 64-74, 2018 02.
Article in English | MEDLINE | ID: mdl-29247852

ABSTRACT

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.


Subject(s)
Cell Differentiation , Cell Plasticity , Macrophages/cytology , Macrophages/physiology , Animals , Biomarkers , Cell Differentiation/immunology , Cell Plasticity/immunology , Disease Susceptibility , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Phenotype
12.
Proc Natl Acad Sci U S A ; 114(50): 13296-13301, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29167373

ABSTRACT

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.


Subject(s)
APOBEC-1 Deaminase/metabolism , Epigenesis, Genetic , RNA Editing , Transcriptome , APOBEC-1 Deaminase/genetics , Animals , Cell Movement , Cells, Cultured , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Phagocytosis
13.
FEBS J ; 284(15): 2410-2424, 2017 08.
Article in English | MEDLINE | ID: mdl-28605567

ABSTRACT

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.


Subject(s)
3' Untranslated Regions , Cell Differentiation , Lymphoma, B-Cell/metabolism , MicroRNAs/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Up-Regulation , Animals , Arginase/genetics , Arginase/metabolism , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation, Neoplastic , Granulocyte Precursor Cells/immunology , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , Immunosuppression Therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , MicroRNAs/antagonists & inhibitors , Monocyte-Macrophage Precursor Cells/immunology , Monocyte-Macrophage Precursor Cells/metabolism , Monocyte-Macrophage Precursor Cells/pathology , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Spleen/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics
14.
Immunity ; 46(5): 835-848.e4, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514689

ABSTRACT

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.


Subject(s)
Cell Differentiation , Clonal Evolution , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Monocytes/cytology , Monocytes/metabolism , Animals , Antigens, CD/metabolism , Biomarkers , Cell Cycle , Cell Lineage , Cell Proliferation , Cells, Cultured , Cluster Analysis , Cytokines/metabolism , Fetal Blood/cytology , Gene Expression Profiling , Humans , Immunophenotyping , Mice
15.
Immunity ; 46(5): 849-862.e7, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514690

ABSTRACT

Monocytes are circulating, short-lived mononuclear phagocytes, which in mice and man comprise two main subpopulations. Murine Ly6C+ monocytes display developmental plasticity and are recruited to complement tissue-resident macrophages and dendritic cells on demand. Murine vascular Ly6C- monocytes patrol the endothelium, act as scavengers, and support vessel wall repair. Here we characterized population and single cell transcriptomes, as well as enhancer and promoter landscapes of the murine monocyte compartment. Single cell RNA-seq and transplantation experiments confirmed homeostatic default differentiation of Ly6C+ into Ly6C- monocytes. The main two subsets were homogeneous, but linked by a more heterogeneous differentiation intermediate. We show that monocyte differentiation occurred through de novo enhancer establishment and activation of pre-established (poised) enhancers. Generation of Ly6C- monocytes involved induction of the transcription factor C/EBPß and C/EBPß-deficient mice lacked Ly6C- monocytes. Mechanistically, C/EBPß bound the Nr4a1 promoter and controlled expression of this established monocyte survival factor.


Subject(s)
Antigens, Ly/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Genomics , Monocytes/metabolism , Animals , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cluster Analysis , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Genomics/methods , High-Throughput Nucleotide Sequencing , Immunophenotyping , Male , Mice , Mice, Knockout , Monocyte-Macrophage Precursor Cells/classification , Monocyte-Macrophage Precursor Cells/metabolism , Monocytes/cytology , Monocytes/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenotype , Promoter Regions, Genetic , Protein Binding
16.
Biomed Res Int ; 2017: 2690402, 2017.
Article in English | MEDLINE | ID: mdl-28286757

ABSTRACT

Objective. Tumor necrosis factor (TNF) increases circulating osteoclast (OC) precursors numbers by promoting their proliferation and differentiation. The aim of this study was to assess the effect of TNF inhibitors (TNFi) on the differentiation and activity of OC in rheumatoid arthritis (RA) patients. Methods. Seventeen RA patients treated with TNFi were analyzed at baseline and after a minimum follow-up period of 6 months. Blood samples were collected to assess receptor activator of nuclear factor kappa-B ligand (RANKL) surface expression on circulating leukocytes and frequency and phenotype of monocyte subpopulations. Quantification of serum levels of bone turnover markers, in vitro OC differentiation assays, and qRT-PCR for OC specific genes was performed. Results. After TNFi therapy, patients had reduced RANKL surface expression in B-lymphocytes and the frequency of circulating classical CD14brightCD16- monocytes was decreased. Serum levels of sRANKL, sRANKL/OPG ratio, and CTX-I were reduced in RA patients after TNFi treatment. Moreover, after exposure to TNFi, osteoclast differentiation and activity were decreased, as well as the expression of TRAF6 and cathepsin K. Conclusion. We propose that TNFi arrests bone loss and erosion, through two pathways: direct reduction of osteoclast precursor numbers and inhibition of intracellular signaling pathways acting through TRAF6.


Subject(s)
Arthritis, Rheumatoid , Monocyte-Macrophage Precursor Cells , Osteoclasts , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cathepsin K/biosynthesis , Female , Follow-Up Studies , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Monocyte-Macrophage Precursor Cells/metabolism , Monocyte-Macrophage Precursor Cells/pathology , Monocytes/metabolism , Monocytes/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/biosynthesis , TNF Receptor-Associated Factor 6/biosynthesis
17.
Endocrinology ; 157(10): 3779-3787, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27471912

ABSTRACT

Fibrocytes are monocyte progenitor cells that have been implicated in normal and pathological tissue remodeling. Among the prominent chemokine receptors expressed by these cells is CXC motif receptor 4 (CXCR4), which, with its cognate ligand CXCL motif ligand 12 (CXCL-12), directs fibrocytes to sites of fibrosis. Fibrocytes have been implicated in the pathogenesis of thyroid-associated ophthalmopathy, the ocular manifestation of Graves' disease (GD), by virtue of their unique accumulation as CD34+ orbital fibroblasts (OFs). Fibrocytes also express high levels of functional TSH receptor (TSHR). Here, we determined CXCL-12 and CXCR4 expression in fibrocytes and GD-OF and whether that pathway interacts with TSHR. CXCL-12 is highly expressed in GD-OF, whereas CXCR4 levels are dramatically higher in fibrocytes. Levels of these proteins are differentially regulated by TSH in a cell type-specific manner. Further, CXCL-12 enhances the induction by TSH of IL-6 in fibrocytes but attenuates this induction in GD-OF. In contrast, in pure CD34+ OF, the interplay between TSH and CXCL-12 reverts to that observed in fibrocytes. Our results indicate that CXCL-12 enhances TSH actions in fibrocytes but inhibits them in GD-OF, a dichotomy imposed by factors emanating from CD34- OF. They also suggest a potentially important modulatory role for CD34- OF in determining the factors that influence pathological TSHR signaling in the TAO orbit.


Subject(s)
Chemokine CXCL12/metabolism , Graves Ophthalmopathy/metabolism , Monocyte-Macrophage Precursor Cells/metabolism , Receptors, CXCR4/metabolism , Receptors, Thyrotropin/metabolism , Cells, Cultured , Humans , Interleukin-6/metabolism , Receptor Cross-Talk
18.
Free Radic Biol Med ; 91: 93-104, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26689473

ABSTRACT

Cholesterol oxidation products such as oxysterols are considered critical factors in the atherosclerotic plaque formation since they induce oxidative stress, inflammation and apoptotic cell death. 27-hydroxycholesterol (27-OH) is one of the most represented oxysterols in atherosclerotic lesions. We recently showed that relatively low concentrations of 27-OH generated a strong survival signaling through an early and transient increase of cellular ROS level, that enhanced MEK-ERK/PI3K-Akt phosphorylation, in turn responsible of a sustained quenching of ROS production. It remains to identify the link between ERK/Akt up-regulation and the consequent quenching effect on ROS intracellular level that efficiently and markedly delay the pro-apoptotic effect of the oxysterol. Here we report on the potent activation of Nrf2 redox-sensitive transcription factor by low micromolar amount of 27-OH added to U937 promonocytic cells. The 27-OH-exerted induction of Nrf2 and subsequently of the target genes, HO-1 and NQO-1, was proved to be: (i) dependent upon the activation of ERK and Akt pathways, (ii) directly responsible for the quenching of intracellular oxidative stress and by this way (iii) ultimately responsible for the observed oxysterol-induced pro-survival response.


Subject(s)
Hydroxycholesterols/pharmacology , Monocyte-Macrophage Precursor Cells/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Apoptosis , Cell Line , Cell Survival , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , MAP Kinase Signaling System , Monocyte-Macrophage Precursor Cells/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species
19.
Naunyn Schmiedebergs Arch Pharmacol ; 389(1): 87-101, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26475619

ABSTRACT

Histamine (HA) is approved for the treatment of acute myeloid leukemia (AML). Its antileukemic activity is related to histamine H2-receptor (H2R)-mediated inhibition of reactive oxygen species (ROS) production in myeloid cells facilitating survival of antineoplastic lymphocytes. The phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, which plays a crucial role in cell survival and proliferation, is constitutively activated in leukemic cells of most AML patients resulting in poor survival prognosis. In a proof-of-principle experiment using a human phosphorylated mitogen-activated protein kinase (MAPK) array, we found high phosphorylation levels of Akt2 in U937 promonocytes that was abrogated by HA or selective H2R agonists. The H2R and the ß2-adrenergic receptor (ß2AR) are Gs-protein-coupled receptors. Stimulation results in adenylyl cyclase activation followed by generation of the second messenger adenosine 3',5'-cyclic monophosphate (cAMP). In our present study, we evaluated the pharmacological profile of the H2R and the ß2AR regarding Akt2 phosphorylation at Ser474 via western blot analysis and ELISA and cAMP accumulation via HPLC-MS/MS in U937 promonocytes. H2R and ß2AR agonists concentration-dependently decreased Akt2 phosphorylation at Ser474. Deviations of potencies and efficacies of agonists in Akt2 phosphorylation and cAMP accumulation assays indicated participation of cAMP-independent signaling in GPCR-induced reduction of Akt2 phosphorylation. Accordingly, our study supports the concept of functional selectivity of the H2R and the ß2AR in U937 promonocytes. In summary, we extended the antileukemic mechanism of HA via H2R and revealed the potential of ß2AR agonists, which are already approved in the treatment of bronchial asthma and chronic obstructive pulmonary disease, as antileukemic drugs.


Subject(s)
Monocyte-Macrophage Precursor Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Histamine H2/metabolism , Cyclic AMP/metabolism , Histamine/pharmacology , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , U937 Cells
20.
Epigenetics ; 10(1): 6-18, 2015.
Article in English | MEDLINE | ID: mdl-25494542

ABSTRACT

Altered expression and activity of histone deacetylases (HDACs) have been correlated with tumorigenesis. Inhibitors of HDACs (HDACi) induce acetylation of histone and non-histone proteins affecting gene expression, cell cycle progression, cell migration, terminal differentiation and cell death. Here, we analyzed the regulation of ARHGEF3, a RhoA-specific guanine nucleotide exchange factor, by the HDACi MS275 (entinostat). MS275 is a well-known benzamide-based HDACi, which induces differentiation of the monoblastic-like human histiocytic lymphoma cell line U937 to monocytes/macrophages. Incubation of U937 cells with MS275 resulted in an up regulation of ARHGEF3, followed by a significant enhancement of the marker of macrophage differentiation CD68. ARHGEF3 protein is primarily nuclear, but MS275 treatment rapidly induced its translocation into the cytoplasm. ARHGEF3 cytoplasmic localization is associated with activation of the RhoA/Rho-associated Kinase (ROCK) pathway. In addition to cytoskeletal rearrangements orchestrated by RhoA, we showed that ARHGEF3/RhoA-dependent signals involve activation of SAPK/JNK and then Elk1 transcription factor. Importantly, MS275-induced CD68 expression was blocked by exposure of U937 cells to exoenzyme C3 transferase and Y27632, inhibitors of Rho and ROCK respectively. Moreover, ARHGEF3 silencing prevented RhoA activation leading to a reduction in SAPK/JNK phosphorylation, Elk1 activation and CD68 expression, suggesting a crucial role for ARHGEF3 in myeloid differentiation. Taken together, our results demonstrate that ARHGEF3 modulates acute myeloid leukemia differentiation through activation of RhoA and pathways directly controlled by small GTPase family proteins. The finding that GEF protein modulation by HDAC inhibition impacts on cell differentiation may be important for understanding the antitumor mechanism(s) by which HDACi treatment stimulates differentiation in cancer.


Subject(s)
Histone Deacetylases/metabolism , Leukemia, Myeloid, Acute/metabolism , Myelopoiesis , Rho Guanine Nucleotide Exchange Factors/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...