Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Ann N Y Acad Sci ; 1499(1): 18-41, 2021 09.
Article in English | MEDLINE | ID: mdl-32445205

ABSTRACT

Tumorigenesis is not only determined by the intrinsic properties of cancer cells but also by their interactions with components of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are among the most abundant immune cells in the TME. During initial stages of tumor development, macrophages can either directly promote antitumor responses by killing tumor cells or indirectly recruit and activate other immune cells. As genetic changes occur within the tumor or T helper 2 (TH 2) cells begin to dominate the TME, TAMs begin to exhibit an immunosuppressive protumor phenotype that promotes tumor progression, metastasis, and resistance to therapy. Thus, targeting TAMs has emerged as a strategy for cancer therapy. To date, TAM targeting strategies have focused on macrophage depletion and inhibition of their recruitment into the TME. However, these strategies have shown limited therapeutic efficacy, although trials are still underway with combination therapies. The fact that macrophages have the potential for antitumor activity has moved the TAM targeting field toward the development of TAM-reprogramming strategies to support this antitumor immune response. Here, we discuss the various roles of TAMs in cancer therapy and their immunosuppressive properties, as well as implications for emerging checkpoint inhibitor-based immunotherapies. We review state-of-the-art TAM-targeting strategies, focusing on current ones at the preclinical and clinical trial stages that aim to reprogram TAMs as an oncological therapy.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Biomarkers, Tumor , Cytotoxicity, Immunologic , Disease Management , Disease Susceptibility , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Blood ; 136(25): 2893-2904, 2020 12 17.
Article in English | MEDLINE | ID: mdl-32614947

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) develop in distinct waves at various anatomical sites during embryonic development. The in vitro differentiation of human pluripotent stem cells (hPSCs) recapitulates some of these processes; however, it has proven difficult to generate functional hematopoietic stem cells (HSCs). To define the dynamics and heterogeneity of HSPCs that can be generated in vitro from hPSCs, we explored single-cell RNA sequencing (scRNAseq) in combination with single-cell protein expression analysis. Bioinformatics analyses and functional validation defined the transcriptomes of naïve progenitors and erythroid-, megakaryocyte-, and leukocyte-committed progenitors, and we identified CD44, CD326, ICAM2/CD9, and CD18, respectively, as markers of these progenitors. Using an artificial neural network that we trained on scRNAseq derived from human fetal liver, we identified a wide range of hPSC-derived HSPCs phenotypes, including a small group classified as HSCs. This transient HSC-like population decreased as differentiation proceeded, and was completely missing in the data set that had been generated using cells selected on the basis of CD43 expression. By comparing the single-cell transcriptome of in vitro-generated HSC-like cells with those generated within the fetal liver, we identified transcription factors and molecular pathways that can be explored in the future to improve the in vitro production of HSCs.


Subject(s)
Antigens, Differentiation , Hematopoietic Stem Cells , Machine Learning , Pluripotent Stem Cells , RNA-Seq , Single-Cell Analysis , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Fetus/cytology , Fetus/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Liver/cytology , Liver/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
3.
J Vis Exp ; (158)2020 04 16.
Article in English | MEDLINE | ID: mdl-32364544

ABSTRACT

Macrophages are present in most vertebrate tissues and comprise widely dispersed and heterogeneous cell populations with different functions. They are key players in health and disease, acting as phagocytes during immune defense and mediating trophic, maintenance, and repair functions. Although it has been possible to study some of the molecular processes involved in human macrophage function, it has proved difficult to apply genetic engineering techniques to primary human macrophages. This has significantly hampered our ability to interrogate the complex genetic pathways involved in macrophage biology and to generate models for specific disease states. An off-the-shelf source of human macrophages that is amenable to the vast arsenal of genetic manipulation techniques would, therefore, provide a valuable tool in this field. We present an optimized protocol that allows for the generation of macrophages from human induced pluripotent stem cells (iPSCs) in vitro. These iPSC-derived macrophages (iPSC-DMs) express human macrophage cell surface markers, including CD45, 25F9, CD163, and CD169, and our live-cell imaging functional assay demonstrates that they exhibit robust phagocytic activity. Cultured iPSC-DMs can be activated to different macrophage states that display altered gene expression and phagocytic activity by the addition of LPS and IFNg, IL4, or IL10. Thus, this system provides a platform to generate human macrophages carrying genetic alterations that model specific human disease and a source of cells for drug screening or cell therapy to treat these diseases.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Biomarkers/metabolism , Cell Count , Cell Differentiation , Cell Membrane/metabolism , Cell Polarity , Cell Shape , Cells, Cultured , Embryoid Bodies/cytology , Humans , Macrophages/metabolism , Phagocytosis , Phenotype
4.
Methods Enzymol ; 632: 113-131, 2020.
Article in English | MEDLINE | ID: mdl-32000892

ABSTRACT

Tumor-associated macrophages (TAMs) are becoming a promising target for cancer immunotherapy. Significant efforts have been made to study the detrimental role of TAMs both in vivo and in vitro. However, it remains challenging to isolate these macrophages to study their function in human cancers and there is the need to seek alternatives to address these limitations. In this review, we will focus on the three most relevant approaches to obtain in vitro fully differentiated macrophages i.e. peripheral blood, immortalized cell lines such as THP-1 or human induced pluripotent stem cells. We will also provide protocols for the polarization of human macrophages to a TAM-like cells in vitro.


Subject(s)
Tumor-Associated Macrophages/cytology , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cell Separation/methods , Humans , Immunophenotyping/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Monocytes/cytology , Monocytes/immunology , Tumor-Associated Macrophages/immunology
5.
Cancer Cell ; 35(4): 588-602.e10, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30930117

ABSTRACT

The roles of tumor-associated macrophages (TAMs) and circulating monocytes in human cancer are poorly understood. Here, we show that monocyte subpopulation distribution and transcriptomes are significantly altered by the presence of endometrial and breast cancer. Furthermore, TAMs from endometrial and breast cancers are transcriptionally distinct from monocytes and their respective tissue-resident macrophages. We identified a breast TAM signature that is highly enriched in aggressive breast cancer subtypes and associated with shorter disease-specific survival. We also identified an auto-regulatory loop between TAMs and cancer cells driven by tumor necrosis factor alpha involving SIGLEC1 and CCL8, which is self-reinforcing through the production of CSF1. Together these data provide direct evidence that monocyte and macrophage transcriptional landscapes are perturbed by cancer, reflecting patient outcomes.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cellular Reprogramming , Macrophages/metabolism , Monocytes/metabolism , Paracrine Communication , Transcription, Genetic , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemokine CCL8/genetics , Chemokine CCL8/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Induced Pluripotent Stem Cells/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophages/pathology , Molecular Targeted Therapy , Monocytes/pathology , Sialic Acid Binding Ig-like Lectin 1/genetics , Sialic Acid Binding Ig-like Lectin 1/metabolism , Signal Transduction , THP-1 Cells , Tumor Microenvironment
6.
Nat Commun ; 10(1): 881, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787325

ABSTRACT

Red blood cells mature within the erythroblastic island (EI) niche that consists of specialized macrophages surrounded by differentiating erythroblasts. Here we establish an in vitro system to model the human EI niche using macrophages that are derived from human induced pluripotent stem cells (iPSCs), and are also genetically programmed to an EI-like phenotype by inducible activation of the transcription factor, KLF1. These EI-like macrophages increase the production of mature, enucleated erythroid cells from umbilical cord blood derived CD34+ haematopoietic progenitor cells and iPSCs; this enhanced production is partially retained even when the contact between progenitor cells and macrophages is inhibited, suggesting that KLF1-induced secreted proteins may be involved in this enhancement. Lastly, we find that the addition of three secreted factors, ANGPTL7, IL-33 and SERPINB2, significantly enhances the production of mature enucleated red blood cells. Our study thus contributes to the ultimate goal of replacing blood transfusion with a manufactured product.


Subject(s)
Erythroblasts/cytology , Erythrocytes/cytology , Erythropoiesis/physiology , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Transcription Factors/metabolism , Macrophages/cytology , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins/metabolism , Antigens, CD34/metabolism , Blood Substitutes/therapeutic use , Blood Transfusion , Hematopoietic Stem Cells/cytology , Humans , Interleukin-33/metabolism , Kruppel-Like Transcription Factors/genetics , Plasminogen Activator Inhibitor 2/metabolism
7.
Article in English | MEDLINE | ID: mdl-29786554

ABSTRACT

We describe the production of a human induced pluripotent stem cell (iPSC) line, SFCi55-ZsGr, that has been engineered to express the fluorescent reporter gene, ZsGreen, in a constitutive manner. The CAG-driven ZsGreen expression cassette was inserted into the AAVS1 locus and a high level of expression was observed in undifferentiated iPSCs and in cell lineages derived from all three germ layers including haematopoietic cells, hepatocytes and neurons. We demonstrate efficient production of terminally differentiated macrophages from the SFCi55-ZsGreen iPSC line and show that they are indistinguishable from those generated from their parental SFCi55 iPSC line in terms of gene expression, cell surface marker expression and phagocytic activity. The high level of ZsGreen expression had no effect on the ability of macrophages to be activated to an M(LPS + IFNγ), M(IL10) or M(IL4) phenotype nor on their plasticity, assessed by their ability to switch from one phenotype to another. Thus, targeting of the AAVS1 locus in iPSCs allows for the production of fully functional, fluorescently tagged human macrophages that can be used for in vivo tracking in disease models. The strategy also provides a platform for the introduction of factors that are predicted to modulate and/or stabilize macrophage function.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.


Subject(s)
Cell Differentiation , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Induced Pluripotent Stem Cells/physiology , Macrophages/metabolism , Cell Lineage/physiology , Germ Layers/growth & development , Humans
8.
Sci Rep ; 7(1): 6394, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743878

ABSTRACT

We have generated a drug-free, all-in-one dCAS9-SAM vector that can activate endogenous gene expression with the potential to modify cell fate. We demonstrate that this strategy can be used in a number of cell lines and avoids exceptionally high levels of gene expression that are observed in standard transgenic approaches. Compared to the multi-plasmid system, this all-in-one vector activates gene expression to a comparable level but the reduced overall DNA content results in significantly higher viability of transfected cells. This allowed us to use the RUNX1C-GFP human embryonic stem cell reporter cell line to monitor gene activation in individual cells and to show that activation could occur at all stages of the cell cycle.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Green Fluorescent Proteins/genetics , Transcriptional Activation , Animals , CRISPR-Cas Systems , Core Binding Factor Alpha 2 Subunit/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Human Embryonic Stem Cells/metabolism , Humans , Mice , RNA, Guide, Kinetoplastida/genetics , Recombinant Proteins/metabolism
9.
PLoS One ; 12(2): e0171698, 2017.
Article in English | MEDLINE | ID: mdl-28212417

ABSTRACT

Engineered nucleases have been used to generate knockout or reporter cell lines and a range of animal models for human disease. These new technologies also hold great promise for therapeutic genome editing. Current methods to evaluate the activity of these nucleases are time consuming, require extensive optimization and are hampered by readouts with low signals and high background. We have developed a simple and easy to perform method (SplitAx) that largely addresses these issues and provides a readout of nuclease activity. The assay involves splitting the N-terminal (amino acid 1-158) coding region of GFP and an out-of-frame of C-terminal region with a nuclease binding site sequence. Following exposure to the test nuclease, cutting and repair by error prone non-homologous end joining (NHEJ) restores the reading frame resulting in the production of a full length fluorescent GFP protein. Fluorescence can also be restored by complementation between the N-terminal and C-terminal coding sequences in trans. We demonstrate successful use of the SplitAx assay to assess the function of zinc finger nucleases, CRISPR hCAS9 and TALENS. We also test the activity of multiple gRNAs in CRISPR/hCas9/D10A systems. The zinc finger nucleases and guide RNAs that showed functional activity in the SplitAx assay were then used successfully to target the endogenous AAVS1, SOX6 and Cfms loci. This simple method can be applied to other unrelated proteins such as ZsGreen1 and provides a test system that does not require complex optimization.


Subject(s)
Endonucleases/genetics , Endonucleases/metabolism , Enzyme Assays/methods , Protein Engineering , Base Sequence , Frameshift Mutation , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...