Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res Commun ; 4(2): 588-606, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38358352

ABSTRACT

Neutrophils are a highly heterogeneous cellular population. However, a thorough examination of the different transcriptional neutrophil states between health and malignancy has not been performed. We utilized single-cell RNA sequencing of human and murine datasets, both publicly available and independently generated, to identify neutrophil transcriptomic subtypes and developmental lineages in health and malignancy. Datasets of lung, breast, and colorectal cancer were integrated to establish and validate neutrophil gene signatures. Pseudotime analysis was used to identify genes driving neutrophil development from health to cancer. Finally, ligand-receptor interactions and signaling pathways between neutrophils and other immune cell populations in primary colorectal cancer and metastatic colorectal cancer were investigated. We define two main neutrophil subtypes in primary tumors: an activated subtype sharing the transcriptomic signatures of healthy neutrophils; and a tumor-specific subtype. This signature is conserved in murine and human cancer, across different tumor types. In colorectal cancer metastases, neutrophils are more heterogeneous, exhibiting additional transcriptomic subtypes. Pseudotime analysis implicates IL1ß/CXCL8/CXCR2 axis in the progression of neutrophils from health to cancer and metastasis, with effects on T-cell effector function. Functional analysis of neutrophil-tumoroid cocultures and T-cell proliferation assays using orthotopic metastatic mouse models lacking Cxcr2 in neutrophils support our transcriptional analysis. We propose that the emergence of metastatic-specific neutrophil subtypes is driven by the IL1ß/CXCL8/CXCR2 axis, with the evolution of different transcriptomic signals that impair T-cell function at the metastatic site. Thus, a better understanding of neutrophil transcriptomic programming could optimize immunotherapeutic interventions into early and late interventions, targeting different neutrophil states. SIGNIFICANCE: We identify two recurring neutrophil populations and demonstrate their staged evolution from health to malignancy through the IL1ß/CXCL8/CXCR2 axis, allowing for immunotherapeutic neutrophil-targeting approaches to counteract immunosuppressive subtypes that emerge in metastasis.


Subject(s)
Colorectal Neoplasms , Neutrophils , Animals , Mice , Humans , Neoplasm Recurrence, Local/metabolism , Signal Transduction/genetics , Colorectal Neoplasms/genetics , Single-Cell Analysis
2.
Gut ; 71(12): 2502-2517, 2022 12.
Article in English | MEDLINE | ID: mdl-35477539

ABSTRACT

OBJECTIVE: Stroma-rich tumours represent a poor prognostic subtype in stage II/III colon cancer (CC), with high relapse rates and limited response to standard adjuvant chemotherapy. DESIGN: To address the lack of efficacious therapeutic options for patients with stroma-rich CC, we stratified our human tumour cohorts according to stromal content, enabling identification of the biology underpinning relapse and potential therapeutic vulnerabilities specifically within stroma-rich tumours that could be exploited clinically. Following human tumour-based discovery and independent clinical validation, we use a series of in vitro and stroma-rich in vivo models to test and validate the therapeutic potential of elevating the biology associated with reduced relapse in human tumours. RESULTS: By performing our analyses specifically within the stroma-rich/high-fibroblast (HiFi) subtype of CC, we identify and validate the clinical value of a HiFi-specific prognostic signature (HPS), which stratifies tumours based on STAT1-related signalling (High-HPS v Low-HPS=HR 0.093, CI 0.019 to 0.466). Using in silico, in vitro and in vivo models, we demonstrate that the HPS is associated with antigen processing and presentation within discrete immune lineages in stroma-rich CC, downstream of double-stranded RNA and viral response signalling. Treatment with the TLR3 agonist poly(I:C) elevated the HPS signalling and antigen processing phenotype across in vitro and in vivo models. In an in vivo model of stroma-rich CC, poly(I:C) treatment significantly increased systemic cytotoxic T cell activity (p<0.05) and reduced liver metastases (p<0.0002). CONCLUSION: This study reveals new biological insight that offers a novel therapeutic option to reduce relapse rates in patients with the worst prognosis CC.


Subject(s)
Biomarkers, Tumor , Colonic Neoplasms , Humans , Biomarkers, Tumor/genetics , Stromal Cells/pathology , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/pathology , Colonic Neoplasms/pathology , Prognosis
3.
Gut ; 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35477863

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is increasingly associated with non-alcoholic steatohepatitis (NASH). HCC immunotherapy offers great promise; however, recent data suggests NASH-HCC may be less sensitive to conventional immune checkpoint inhibition (ICI). We hypothesised that targeting neutrophils using a CXCR2 small molecule inhibitor may sensitise NASH-HCC to ICI therapy. DESIGN: Neutrophil infiltration was characterised in human HCC and mouse models of HCC. Late-stage intervention with anti-PD1 and/or a CXCR2 inhibitor was performed in murine models of NASH-HCC. The tumour immune microenvironment was characterised by imaging mass cytometry, RNA-seq and flow cytometry. RESULTS: Neutrophils expressing CXCR2, a receptor crucial to neutrophil recruitment in acute-injury, are highly represented in human NASH-HCC. In models of NASH-HCC lacking response to ICI, the combination of a CXCR2 antagonist with anti-PD1 suppressed tumour burden and extended survival. Combination therapy increased intratumoural XCR1+ dendritic cell activation and CD8+ T cell numbers which are associated with anti-tumoural immunity, this was confirmed by loss of therapeutic effect on genetic impairment of myeloid cell recruitment, neutralisation of the XCR1-ligand XCL1 or depletion of CD8+ T cells. Therapeutic benefit was accompanied by an unexpected increase in tumour-associated neutrophils (TANs) which switched from a protumour to anti-tumour progenitor-like neutrophil phenotype. Reprogrammed TANs were found in direct contact with CD8+ T cells in clusters that were enriched for the cytotoxic anti-tumoural protease granzyme B. Neutrophil reprogramming was not observed in the circulation indicative of the combination therapy selectively influencing TANs. CONCLUSION: CXCR2-inhibition induces reprogramming of the tumour immune microenvironment that promotes ICI in NASH-HCC.

4.
Cancer Cell ; 36(3): 319-336.e7, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31526760

ABSTRACT

The metastatic process of colorectal cancer (CRC) is not fully understood and effective therapies are lacking. We show that activation of NOTCH1 signaling in the murine intestinal epithelium leads to highly penetrant metastasis (100% metastasis; with >80% liver metastases) in KrasG12D-driven serrated cancer. Transcriptional profiling reveals that epithelial NOTCH1 signaling creates a tumor microenvironment (TME) reminiscent of poorly prognostic human CRC subtypes (CMS4 and CRIS-B), and drives metastasis through transforming growth factor (TGF) ß-dependent neutrophil recruitment. Importantly, inhibition of this recruitment with clinically relevant therapeutic agents blocks metastasis. We propose that NOTCH1 signaling is key to CRC progression and should be exploited clinically.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Receptor, Notch1/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Datasets as Topic , Disease Models, Animal , Gene Expression Profiling , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Male , Mice , Mutation , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Neutrophils/immunology , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Notch1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Survival Analysis , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
5.
Stem Cell Reports ; 10(1): 151-165, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29276152

ABSTRACT

In vivo hematopoietic generation occurs in waves of primitive and definitive cell emergence. Differentiation cultures of pluripotent embryonic stem cells (ESCs) offer an accessible source of hematopoietic cells for blood-related research and therapeutic strategies. However, despite many approaches, it remains a goal to robustly generate hematopoietic progenitor and stem cells (HP/SCs) in vitro from ESCs. This is partly due to the inability to efficiently promote, enrich, and/or molecularly direct hematopoietic emergence. Here, we use Gata2Venus (G2V) and Ly6a(SCA1)GFP (LG) reporter ESCs, derived from well-characterized mouse models of HP/SC emergence, to show that during in vitro differentiation they report emergent waves of primitive hematopoietic progenitor cells (HPCs), definitive HPCs, and B-lymphoid cell potential. These results, facilitated by enrichment of single and double reporter cells with HPC properties, demonstrate that in vitro ESC differentiation approximates the waves of hematopoietic cell generation found in vivo, thus raising possibilities for enrichment of rare ESC-derived HP/SCs.


Subject(s)
Cell Differentiation , GATA2 Transcription Factor , Genes, Reporter , Hematopoietic Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology
6.
Biochim Biophys Acta Gene Regul Mech ; 1860(6): 721-729, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28391050

ABSTRACT

Transcriptional activation of the EVI1 oncogene (3q26) leads to aggressive forms of human acute myeloid leukemia (AML). However, the mechanism of EVI1-mediated leukemogenesis has not been fully elucidated. Previously, by characterizing the EVI1 promoter, we have shown that RUNX1 and ELK1 directly regulate EVI1 transcription. Intriguingly, bioinformatic analysis of the EVI1 promoter region identified the presence of several EVI1 potential binding sites. Thus, we hypothesized that EVI1 could bind to these sites regulating its own transcription. In this study, we show that there is a functional interaction between EVI1 and its promoter, and that the different EVI1 isoforms (EVI1-145kDa, EVI1-Δ324 and MDS1-EVI1) regulate the transcription of EVI1 transcripts through distinct promoter regions. Moreover, we determine that the EVI1-145kDa isoform activates EVI1 transcription, whereas EVI1-Δ324 and MDS1-EVI1 act as repressors. Finally, we demonstrate that these EVI1 isoforms are involved in cell transformation; functional experiments show that EVI1-145kDa prolongs the maintenance of hematopoietic stem and progenitor cells; conversely, MDS1-EVI1 repressed hematopoietic stem and progenitor colony replating capacity. We demonstrate for the first time that EVI1 acts as a regulator of its own expression, highlighting the complex regulation of EVI1, and open new directions to better understand the mechanisms of EVI1 overexpressing leukemias.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/pathology , Humans , Leukemia/genetics , Leukemia/pathology , MDS1 and EVI1 Complex Locus Protein , Mice , Proto-Oncogenes/genetics , Transcription Factors/genetics
7.
J Immunol ; 194(5): 2190-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25624456

ABSTRACT

Germline GATA2 mutations have been identified as the cause of familial syndromes with immunodeficiency and predisposition to myeloid malignancies. GATA2 mutations appear to cause loss of function of the mutated allele leading to haploinsufficiency; however, this postulate has not been experimentally validated as the basis of these syndromes. We hypothesized that mutations that are translated into abnormal proteins could affect the transcription of GATA2, triggering GATA2 deficiency. Chromatin immunoprecipitation and luciferase assays showed that the human GATA2 protein activates its own transcription through a specific region located at -2.4 kb, whereas the p.Thr354Met, p.Thr355del, and p.Arg396Gln germline mutations impair GATA2 promoter activation. Accordingly, GATA2 expression was decreased to ∼58% in a patient with p.Arg396Gln, compared with controls. p.Arg396Gln is the second most common mutation in these syndromes, and no previous functional analyses have been performed. We therefore analyzed p.Arg396Gln. Our data show that p.Arg396Gln is a loss-of-function mutation affecting DNA-binding ability and, as a consequence, it fails to maintain the immature characteristics of hematopoietic stem and progenitor cells, which could result in defects in this cell compartment. In conclusion, we show that human GATA2 binds to its own promoter, activating its transcription, and that the aforementioned mutations impair the transcription of GATA2. Our results indicate that they can affect other GATA2 target genes, which could partially explain the variability of symptoms in these diseases. Moreover, we show that p.Arg396Gln is a loss-of-function mutation, which is unable to retain the progenitor phenotype in cells where it is expressed.


Subject(s)
GATA2 Transcription Factor/genetics , Germ-Line Mutation , Immunologic Deficiency Syndromes/immunology , Mycobacterium avium-intracellulare Infection/immunology , Transcription, Genetic , Alleles , Binding Sites , Cell Line, Tumor , Female , GATA2 Transcription Factor/immunology , Gene Expression Regulation , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Middle Aged , Models, Molecular , Mycobacterium avium Complex/immunology , Mycobacterium avium-intracellulare Infection/complications , Mycobacterium avium-intracellulare Infection/genetics , Mycobacterium avium-intracellulare Infection/pathology , Phenotype , Promoter Regions, Genetic , Protein Binding , Signal Transduction
8.
Haematologica ; 97(4): 543-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22133779

ABSTRACT

BACKGROUND: Protein phosphatase 2A is a novel potential therapeutic target in several types of chronic and acute leukemia, and its inhibition is a common event in acute myeloid leukemia. Upregulation of SET is essential to inhibit protein phosphatase 2A in chronic myeloid leukemia, but its importance in acute myeloid leukemia has not yet been explored. DESIGN AND METHODS: We quantified SET expression by real time reverse transcriptase polymerase chain reaction in 214 acute myeloid leukemia patients at diagnosis. Western blot was performed in acute myeloid leukemia cell lines and in 16 patients' samples. We studied the effect of SET using cell viability assays. Bioinformatics analysis of the SET promoter, chromatin immunoprecipitation, and luciferase assays were performed to evaluate the transcriptional regulation of SET. RESULTS: SET overexpression was found in 60/214 patients, for a prevalence of 28%. Patients with SET overexpression had worse overall survival (P<0.01) and event-free survival (P<0.01). Deregulation of SET was confirmed by western blot in both cell lines and patients' samples. Functional analysis showed that SET promotes proliferation, and restores cell viability after protein phosphatase 2A overexpression. We identified EVI1 overexpression as a mechanism involved in SET deregulation in acute myeloid leukemia cells. CONCLUSIONS: These findings suggest that SET overexpression is a key mechanism in the inhibition of PP2A in acute myeloid leukemia, and that EVI1 overexpression contributes to the deregulation of SET. Furthermore, SET overexpression is associated with a poor outcome in acute myeloid leukemia, and it can be used to identify a subgroup of patients who could benefit from future treatments based on PP2A activators.


Subject(s)
Gene Expression , Histone Chaperones/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , DNA-Binding Proteins , Female , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Prognosis , Protein Phosphatase 2/antagonists & inhibitors , Recurrence , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...