Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Liver Int ; 43(1): 208-220, 2023 01.
Article in English | MEDLINE | ID: mdl-35924447

ABSTRACT

BACKGROUND AND AIMS: Dickkopf-1 (DKK1) is associated with poor prognosis in intrahepatic cholangiocarcinoma (iCCA), but the mechanisms behind this are unclear. Here, we show that DKK1 plays an immune regulatory role in vivo and inhibition reduces tumour growth. METHODS: Various in vivo GEMM mouse models and patient samples were utilized to assess the effects of tumour specific DKK1 overexpression in iCCA. DKK1-driven changes to the tumour immune microenvironment were characterized by immunostaining and gene expression analysis. DKK1 overexpressing and damage-induced models of iCCA were used to demonstrate the therapeutic efficacy of DKK1 inhibition in these contexts using the anti-DKK1 therapeutic, DKN-01. RESULTS: DKK1 overexpression in mouse models of iCCA drives an increase in chemokine and cytokine signalling, the recruitment of regulatory macrophages, and promotes the formation of a tolerogenic niche with higher numbers of regulatory T cells. We show a similar association of DKK1 with FOXP3 and regulatory T cells in patient tissue and gene expression data, demonstrating these effects are relevant to human iCCA. Finally, we demonstrate that inhibition of DKK1 with the monoclonal antibody mDKN-01 is effective at reducing tumour burden in two distinct mouse models of the disease. CONCLUSION: DKK1 promotes tumour immune evasion in iCCA through the recruitment of immune suppressive macrophages. Targeting DKK1 with a neutralizing antibody is effective at reducing tumour growth in vivo. As such, DKK1 targeted and immune modulatory therapies may be an effective strategy in iCCA patients with high DKK1 tumour expression or tolerogenic immune phenotypes.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Intercellular Signaling Peptides and Proteins , Animals , Humans , Mice , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Disease Models, Animal , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/genetics , Phenotype , Tumor Microenvironment
3.
Cancer Immunol Res ; 7(12): 2052-2064, 2019 12.
Article in English | MEDLINE | ID: mdl-31615815

ABSTRACT

Metastasis-associated macrophages (MAM) promote persistent growth of breast cancer cells at the metastatic site and are, thus, an attractive therapeutic target to treat breast cancer metastasis, a leading cause of cancer-related death in women. However, the precise mechanisms behind MAM-mediated metastatic tumor outgrowth have not been fully elucidated. Using mouse models of metastatic breast cancer, we showed that MAMs uniquely expressed hepatocyte growth factor (HGF) in metastatic tumors. We also demonstrated that a selected population of cancer cells with high metastatic potential (cancer cells that can establish metastatic tumors in mice with higher number and incidence than parental cells) had higher expression of HGF receptor, MNNG HOS transforming gene (MET), and were more responsive to HGF released from macrophages compared with the parental cells. Blockade of MET signaling in cancer cells suppressed metastatic tumor expansion, in part, through activation of natural killer cells. Results from this study suggest an approach to prevent life-threatening metastatic tumor formation using blockade of MAM-induced MET signal activation in metastatic cancer cells.


Subject(s)
Hepatocyte Growth Factor/genetics , Macrophages/metabolism , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-met/genetics , Animals , Cell Line, Tumor , Female , Humans , Killer Cells, Natural , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Proto-Oncogene Proteins c-met/metabolism
4.
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
5.
J Exp Med ; 212(9): 1433-48, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26261265

ABSTRACT

Although the link between inflammation and cancer initiation is well established, its role in metastatic diseases, the primary cause of cancer deaths, has been poorly explored. Our previous studies identified a population of metastasis-associated macrophages (MAMs) recruited to the lung that promote tumor cell seeding and growth. Here we show that FMS-like tyrosine kinase 1 (Flt1, also known as VEGFR1) labels a subset of macrophages in human breast cancers that are significantly enriched in metastatic sites. In mouse models of breast cancer pulmonary metastasis, MAMs uniquely express FLT1. Using several genetic models, we show that macrophage FLT1 signaling is critical for metastasis. FLT1 inhibition does not affect MAM recruitment to metastatic lesions but regulates a set of inflammatory response genes, including colony-stimulating factor 1 (CSF1), a central regulator of macrophage biology. Using a gain-of-function approach, we show that CSF1-mediated autocrine signaling in MAMs is downstream of FLT1 and can restore the tumor-promoting activity of FLT1-inhibited MAMs. Thus, CSF1 is epistatic to FLT1, establishing a link between FLT1 and inflammatory responses within breast tumor metastases. Importantly, FLT1 inhibition reduces tumor metastatic efficiency even after initial seeding, suggesting that these pathways represent therapeutic targets in metastatic disease.


Subject(s)
Breast Neoplasms/metabolism , Macrophages/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Autocrine Communication/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Proteins/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics
6.
Curr Protoc Cell Biol ; Chapter 21: Unit 21.2, 2004 Oct.
Article in English | MEDLINE | ID: mdl-18228451

ABSTRACT

Fluorescence localization after photobleaching is a new method for localized photolabeling and subsequent tracking of specific molecules within living cells. The molecular species to be located carries two different fluorophores that can be imaged independently but simultaneously by fluorescence microscopy. For the method to work, these two fluorophores should be accurately colocalized throughout the cell so that their images are closely matched. One of the fluorophores (the target fluorophore) is then rapidly photobleached at a chosen location. The unbleached (reference) fluorophore remains colocalized with the target fluorophore; thus, the subsequent fate of the photobleached molecules can be revealed by processing simultaneously acquired digital images of the two fluorophores. Here we demonstrate the simplicity and effectiveness of the FLAP method in revealing both fast and slow molecular dynamics in living cells using a Zeiss LSM 510 laser scanning confocal microscope.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Animals , Cells, Cultured , Fluorescence Recovery After Photobleaching/instrumentation , Image Processing, Computer-Assisted/methods , Models, Biological , Software
7.
Science ; 300(5616): 142-5, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12677069

ABSTRACT

Transformed rat fibroblasts expressing two variants of green fluorescent protein, each fused to beta-actin, were used to study actin dynamics during cell protrusion. The recently developed FLAP (fluorescence localization after photobleaching) method permits the tracking of one fluorophore after localized photobleaching by using the other as a colocalized reference. Here, by visualizing the ratio of bleached to total molecules, we found that actin was delivered to protruding zones of the leading edge of the cell at speeds that exceeded 5 micrometers per second. Monte Carlo modeling confirmed that this flow cannot be explained by diffusion and may involve active transport.


Subject(s)
Actins/metabolism , Depsipeptides , Pseudopodia/physiology , Pseudopodia/ultrastructure , Amides/pharmacology , Animals , Azepines/pharmacology , Bacterial Proteins/metabolism , Biological Transport, Active , Biopolymers , Cell Line, Transformed , Cell Movement , Diffusion , Enzyme Inhibitors/pharmacology , Fluorescence , Fluorescence Recovery After Photobleaching , Fluorometry , Green Fluorescent Proteins , Image Processing, Computer-Assisted , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Microscopy, Confocal , Monte Carlo Method , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/pharmacology , Nocodazole/pharmacology , Peptides, Cyclic/pharmacology , Photobleaching , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Pseudopodia/drug effects , Pyridines/pharmacology , Rats , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...