Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Cell ; 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39137777

ABSTRACT

Tumors growing in metabolically challenged environments, such as glioblastoma in the brain, are particularly reliant on crosstalk with their tumor microenvironment (TME) to satisfy their high energetic needs. To study the intricacies of this metabolic interplay, we interrogated the heterogeneity of the glioblastoma TME using single-cell and multi-omics analyses and identified metabolically rewired tumor-associated macrophage (TAM) subpopulations with pro-tumorigenic properties. These TAM subsets, termed lipid-laden macrophages (LLMs) to reflect their cholesterol accumulation, are epigenetically rewired, display immunosuppressive features, and are enriched in the aggressive mesenchymal glioblastoma subtype. Engulfment of cholesterol-rich myelin debris endows subsets of TAMs to acquire an LLM phenotype. Subsequently, LLMs directly transfer myelin-derived lipids to cancer cells in an LXR/Abca1-dependent manner, thereby fueling the heightened metabolic demands of mesenchymal glioblastoma. Our work provides an in-depth understanding of the immune-metabolic interplay during glioblastoma progression, thereby laying a framework to unveil targetable metabolic vulnerabilities in glioblastoma.

2.
Nat Immunol ; 23(7): 1031-1041, 2022 07.
Article in English | MEDLINE | ID: mdl-35761082

ABSTRACT

The immune checkpoint receptor lymphocyte activation gene 3 protein (LAG3) inhibits T cell function upon binding to major histocompatibility complex class II (MHC class II) or fibrinogen-like protein 1 (FGL1). Despite the emergence of LAG3 as a target for next-generation immunotherapies, we have little information describing the molecular structure of the LAG3 protein or how it engages cellular ligands. Here we determined the structures of human and murine LAG3 ectodomains, revealing a dimeric assembly mediated by Ig domain 2. Epitope mapping indicates that a potent LAG3 antagonist antibody blocks interactions with MHC class II and FGL1 by binding to a flexible 'loop 2' region in LAG3 domain 1. We also defined the LAG3-FGL1 interface by mapping mutations onto structures of LAG3 and FGL1 and established that FGL1 cross-linking induces the formation of higher-order LAG3 oligomers. These insights can guide LAG3-based drug development and implicate ligand-mediated LAG3 clustering as a mechanism for disrupting T cell activation.


Subject(s)
Antigens, CD/metabolism , Lymphocyte Activation , Animals , Antibodies , Fibrinogen , Histocompatibility Antigens Class II/metabolism , Humans , Immunotherapy , Ligands , Mice , Receptors, Immunologic , Lymphocyte Activation Gene 3 Protein
3.
Immunity ; 55(10): 1761-1763, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223725

ABSTRACT

Lineage plasticity is a critical mechanism of therapeutic resistance in cancer. In a recent issue of Science, Chan and colleagues demonstrate that early lineage plasticity in prostate cancer is driven by JAK-STAT inflammatory cytokine signaling.


Subject(s)
Cytokines , Prostatic Neoplasms , Cytokines/metabolism , Humans , Janus Kinases/metabolism , Male , STAT Transcription Factors/metabolism , Signal Transduction
4.
Immunity ; 54(6): 1154-1167.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33979578

ABSTRACT

Blockade of the inhibitory receptor TIM-3 shows efficacy in cancer immunotherapy clinical trials. TIM-3 inhibits production of the chemokine CXCL9 by XCR1+ classical dendritic cells (cDC1), thereby limiting antitumor immunity in mammary carcinomas. We found that increased CXCL9 expression by splenic cDC1s upon TIM-3 blockade required type I interferons and extracellular DNA. Chemokine expression as well as combinatorial efficacy of TIM-3 blockade and paclitaxel chemotherapy were impaired by deletion of Cgas and Sting. TIM-3 blockade increased uptake of extracellular DNA by cDC1 through an endocytic process that resulted in cytoplasmic localization. DNA uptake and efficacy of TIM-3 blockade required DNA binding by HMGB1, while galectin-9-induced cell surface clustering of TIM-3 was necessary for its suppressive function. Human peripheral blood cDC1s also took up extracellular DNA upon TIM-3 blockade. Thus, TIM-3 regulates endocytosis of extracellular DNA and activation of the cytoplasmic DNA sensing cGAS-STING pathway in cDC1s, with implications for understanding the mechanisms underlying TIM-3 immunotherapy.


Subject(s)
DNA/metabolism , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , Animals , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Chemokines/metabolism , Cytoplasm/metabolism , Endocytosis/physiology , Female , HEK293 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL
5.
Immunity ; 52(4): 668-682.e7, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294407

ABSTRACT

The primary mechanisms supporting immunoregulatory polarization of myeloid cells upon infiltration into tumors remain largely unexplored. Elucidation of these signals could enable better strategies to restore protective anti-tumor immunity. Here, we investigated the role of the intrinsic activation of the PKR-like endoplasmic reticulum (ER) kinase (PERK) in the immunoinhibitory actions of tumor-associated myeloid-derived suppressor cells (tumor-MDSCs). PERK signaling increased in tumor-MDSCs, and its deletion transformed MDSCs into myeloid cells that activated CD8+ T cell-mediated immunity against cancer. Tumor-MDSCs lacking PERK exhibited disrupted NRF2-driven antioxidant capacity and impaired mitochondrial respiratory homeostasis. Moreover, reduced NRF2 signaling in PERK-deficient MDSCs elicited cytosolic mitochondrial DNA elevation and, consequently, STING-dependent expression of anti-tumor type I interferon. Reactivation of NRF2 signaling, conditional deletion of STING, or blockade of type I interferon receptor I restored the immunoinhibitory potential of PERK-ablated MDSCs. Our findings demonstrate the pivotal role of PERK in tumor-MDSC functionality and unveil strategies to reprogram immunosuppressive myelopoiesis in tumors to boost cancer immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/immunology , Carcinoma, Ovarian Epithelial/immunology , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/immunology , Membrane Proteins/immunology , Skin Neoplasms/immunology , eIF-2 Kinase/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Immunosuppression Therapy , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Unfolded Protein Response/immunology , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
6.
Immunity ; 48(1): 6-8, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343441

ABSTRACT

Type 1 conventional dendritic cells are necessary for the development of anti-tumor immunity. In this issue of Immunity, Sharma et al. (2018) identify a phenotypically similar monocyte-derived population within inflamed tumors that promotes T cell responses during therapy.


Subject(s)
Cell Differentiation , Cells, Cultured , Dendritic Cells , Monocytes , T-Lymphocytes
7.
J Immunol ; 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120462

ABSTRACT

Immunotherapy response is associated with the presence of conventional dendritic cells (cDCs). cDC type 1 (cDC1) is critically important for CD8+ T cell activation, cDC type 2 (cDC2) regulates CD4+ T cell responses, and mature regulatory cDCs may dampen T cell responses in the tumor microenvironment (TME). However, we lack a clear understanding of cDC distribution in the human TME, cDC prevalence in metastatic sites, and cDC differences in early- versus late-stage disease. Rapid autopsy specimens of 10 patients with lung adenocarcinoma were evaluated to detect cDCs and immune cells via multiplex immunofluorescence using 18 markers and 42 tumors. First, we found that T cells, cDC1, and cDC2 were confined to stroma, whereas mature regulatory DCs were enriched in tumor, suggesting unique localization-specific functions. Second, lung and lymph node tumors were more enriched in T cells and cDCs than liver tumors, underscoring differences in the TME of metastatic sites. Third, although the proportion of T cells and cDC1 did not differ in different stages, an increase in the proportion of cDC2 and macrophages in late stage suggests potential differences in regulation of T cell responses in different stages. Collectively, these findings provide new, to our knowledge, insights into cDC biology in human cancer that may have important therapeutic implications.

8.
Nat Mater ; 20(4): 548-559, 2021 04.
Article in English | MEDLINE | ID: mdl-33257795

ABSTRACT

Stromal stiffening accompanies malignancy, compromises treatment and promotes tumour aggression. Clarifying the molecular nature and the factors that regulate stromal stiffening in tumours should identify biomarkers to stratify patients for therapy and interventions to improve outcome. We profiled lysyl hydroxylase-mediated and lysyl oxidase-mediated collagen crosslinks and quantified the greatest abundance of total and complex collagen crosslinks in aggressive human breast cancer subtypes with the stiffest stroma. These tissues harbour the highest number of tumour-associated macrophages, whose therapeutic ablation in experimental models reduced metastasis, and decreased collagen crosslinks and stromal stiffening. Epithelial-targeted expression of the crosslinking enzyme, lysyl oxidase, had no impact on collagen crosslinking in PyMT mammary tumours, whereas stromal cell targeting did. Stromal cells in microdissected human tumours expressed the highest level of collagen crosslinking enzymes. Immunohistochemical analysis of biopsies from a cohort of patients with breast cancer revealed that stromal expression of lysyl hydroxylase 2, an enzyme that induces hydroxylysine aldehyde-derived collagen crosslinks and stromal stiffening, correlated significantly with disease specific mortality. The findings link tissue inflammation, stromal cell-mediated collagen crosslinking and stiffening to tumour aggression and identify lysyl hydroxylase 2 as a stromal biomarker.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Collagen/metabolism , Stromal Cells/metabolism , Tumor-Associated Macrophages/metabolism , Adult , Biopsy , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Humans , Middle Aged , Protein-Lysine 6-Oxidase/metabolism , Stromal Cells/pathology
9.
Immunity ; 38(4): 626-8, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601679

ABSTRACT

In this issue of Immunity, Ma et al. (2013) demonstrate that neoplastic cell release of ATP drives recruitment and differentiation of CD11b(+) dendritic cells within tumors, where their ability to locally present antigen improves response to anthracycline-based chemotherapy.

10.
Genes Dev ; 27(19): 2086-98, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24065739

ABSTRACT

Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/physiopathology , Cathepsin C/metabolism , Skin Neoplasms/physiopathology , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin C/genetics , Cell Line, Tumor , Chymases/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukocytes/metabolism , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Pancreatic Elastase/metabolism
12.
J Immunol ; 201(2): 734-746, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29884705

ABSTRACT

The mammary gland is not classically considered a mucosal organ, although it exhibits some features common to mucosal tissues. Notably, the mammary epithelium is contiguous with the external environment, is exposed to bacteria during lactation, and displays antimicrobial features. Nonetheless, immunological hallmarks predictive of mucosal function have not been demonstrated in the mammary gland, including immune tolerance to foreign Ags under homeostasis. This inquiry is important, as mucosal immunity in the mammary gland may assure infant and women's health during lactation. Further, such mucosal immune programs may protect mammary function at the expense of breast cancer promotion via decreased immune surveillance. In this study, using murine models, we evaluated mammary specific mucosal attributes focusing on two reproductive states at increased risk for foreign and self-antigen exposure: lactation and weaning-induced involution. We find a baseline mucosal program of RORγT+ CD4+ T cells that is elevated within lactating and involuting mammary glands and is extended during involution to include tolerogenic dendritic cell phenotypes, barrier-supportive antimicrobials, and immunosuppressive Foxp3+ CD4+ T cells. Further, we demonstrate suppression of Ag-dependent CD4+ T cell activation, data consistent with immune tolerance. We also find Ag-independent accumulation of memory RORγT+ Foxp3+ CD4+ T cells specifically within the involution mammary gland consistent with an active immune process. Overall, these data elucidate strong mucosal immune programs within lactating and involuting mammary glands. Our findings support the classification of the mammary gland as a temporal mucosal organ and open new avenues for exploration into breast pathologic conditions, including compromised lactation and breast cancer.


Subject(s)
Immunity, Mucosal , Mammary Glands, Animal/physiology , Mammary Glands, Human/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Lactation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
13.
Br J Cancer ; 121(7): 556-566, 2019 10.
Article in English | MEDLINE | ID: mdl-31417189

ABSTRACT

BACKGROUND: Tumours rapidly ferment glucose to lactic acid even in the presence of oxygen, and coupling high glycolysis with poor perfusion leads to extracellular acidification. We hypothesise that acidity, independent from lactate, can augment the pro-tumour phenotype of macrophages. METHODS: We analysed publicly available data of human prostate cancer for linear correlation between macrophage markers and glycolysis genes. We used zwitterionic buffers to adjust the pH in series of in vitro experiments. We then utilised subcutaneous and transgenic tumour models developed in C57BL/6 mice as well as computer simulations to correlate tumour progression with macrophage infiltration and to delineate role of acidity. RESULTS: Activating macrophages at pH 6.8 in vitro enhanced an IL-4-driven phenotype as measured by gene expression, cytokine profiling, and functional assays. These results were recapitulated in vivo wherein neutralising intratumoural acidity reduced the pro-tumour phenotype of macrophages, while also decreasing tumour incidence and invasion in the TRAMP model of prostate cancer. These results were recapitulated using an in silico mathematical model that simulate macrophage responses to environmental signals. By turning off acid-induced cellular responses, our in silico mathematical modelling shows that acid-resistant macrophages can limit tumour progression. CONCLUSIONS: This study suggests that tumour acidity contributes to prostate carcinogenesis by altering the state of macrophage activation.


Subject(s)
Disease Progression , Macrophage Activation , Macrophages/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Bicarbonates/pharmacology , Cell Line, Tumor , Computer Simulation , Cytokines/metabolism , Extracellular Space/metabolism , Gene Expression , Glucose/metabolism , Glycolysis/genetics , Humans , Hydrogen-Ion Concentration , Interleukin-4/metabolism , Lactic Acid/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Theoretical , Neoplasm Invasiveness , Phenotype , Random Allocation , Tumor Microenvironment
14.
Trends Immunol ; 37(12): 855-865, 2016 12.
Article in English | MEDLINE | ID: mdl-27793569

ABSTRACT

Dendritic cells (DCs) are central regulators of the adaptive immune response, and as such are necessary for T-cell-mediated cancer immunity. In particular, antitumoral responses depend on a specialized subset of conventional DCs that transport tumor antigens to draining lymph nodes and cross-present antigen to activate cytotoxic T lymphocytes. DC maturation is necessary to provide costimulatory signals to T cells, but while DC maturation occurs within tumors, it is often insufficient to induce potent immunity, particularly in light of suppressive mechanisms within tumors. Bypassing suppressive pathways or directly activating DCs can unleash a T-cell response, and although clinical efficacy has proven elusive, therapeutic targeting of DCs continues to hold translational potential in combinatorial approaches.


Subject(s)
Antigen Presentation , Cancer Vaccines/immunology , Cross-Priming , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Cell Differentiation , Dendritic Cells/transplantation , Humans , Neoplasms/therapy , Tumor Microenvironment
15.
Breast Cancer Res Treat ; 171(2): 283-293, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29858753

ABSTRACT

INTRODUCTION: We hypothesized that breast tissue not involved by tumor in inflammatory breast cancer (IBC) patients contains intrinsic differences, including increased mammary stem cells and macrophage infiltration, which may promote the IBC phenotype. MATERIALS AND METHODS: Normal breast parenchyma ≥ 5 cm away from primary tumors was obtained from mastectomy specimens. This included an initial cohort of 8 IBC patients and 60 non-IBC patients followed by a validation cohort of 19 IBC patients and 25 non-IBC patients. Samples were immunostained for either CD44+CD49f+CD133/2+ mammary stem cell markers or the CD68 macrophage marker and correlated with IBC status. Quantitation of positive cells was determined using inForm software from PerkinElmer. We also examined the association between IBC status and previously published tumorigenic stem cell and IBC tumor signatures in the validation cohort samples. RESULTS: 8 of 8 IBC samples expressed isolated CD44+CD49f+CD133/2+ stem cell marked cells in the initial cohort as opposed to 0/60 non-IBC samples (p = 0.001). Similarly, the median number of CD44+CD49f+CD133/2+ cells was significantly higher in the IBC validation cohort as opposed to the non-IBC validation cohort (25.7 vs. 14.2, p = 0.007). 7 of 8 IBC samples expressed CD68 + histologically confirmed macrophages in initial cohort as opposed to 12/48 non-IBC samples (p = 0.001). In the validation cohort, the median number of CD68 + cells in IBC was 3.7 versus 1.0 in the non-IBC cohort (p = 0.06). IBC normal tissue was positively associated with a tumorigenic stem cell signature (p = 0.02) and with a 79-gene IBC signature (p < 0.001). CONCLUSIONS: Normal tissue from IBC patients is enriched for both mammary stem cells and macrophages and has higher association with both a tumorigenic stem cell signature and IBC-specific tumor signature. Collectively, these data suggest that IBC normal tissue differs from non-IBC tissue. Whether these changes occur before the tumor develops or is induced by tumor warrants further investigation.


Subject(s)
Inflammatory Breast Neoplasms/immunology , Inflammatory Breast Neoplasms/metabolism , Macrophages/immunology , Macrophages/metabolism , Stem Cells/metabolism , Biomarkers , Cell Line, Tumor , Female , Gene Expression , Humans , Immunohistochemistry , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/pathology , Macrophages/pathology , Neoplasm Grading , Neoplasm Staging , Reproducibility of Results
16.
Anticancer Drugs ; 28(1): 120-126, 2017 01.
Article in English | MEDLINE | ID: mdl-27669423

ABSTRACT

The aims of this study were to report the clinical outcomes in a cohort of men with high-risk prostate cancer treated with neoadjuvant docetaxel and mitoxantrone 10 years after treatment, identify pretreatment clinical parameters that may be predictors of recurrence, and describe tumor-infiltrating leukocytes present in radical prostatectomy specimens. We conducted a phase I/II study of neoadjuvant docetaxel and mitoxantrone before radical prostatectomy in high-risk localized prostate cancer to determine the feasibility of this combination and predictors of prostate cancer recurrence after cytotoxic chemotherapy. After 10 years of follow-up, 34 (63%) of 54 participants experience a recurrence. In univariate analysis, prostate-specific antigen (PSA) density (P=0.01), pathological stage (P=0.03), lymph node status (P<0.0001), seminal vesicle invasion (P=0.003), and tissue vascular endothelial growth factor (VEGF) expression (P=0.016) were significantly associated with recurrence. In multivariate analysis, only lymph node status, PSA density, and VEGF expression were significant predictors of disease recurrence. We used a tissue microarray for the first 50 participants to characterize the tumor-infiltrating lymphocytes and evaluate them for association with recurrence. We measured CD3, CD4, CD8, FoxP3, CD20, CD15, CD68, and CD163 by immunohistochemistry in both tumor and normal prostate specimens, but did not find an association between immunophenotype and recurrence. There was a significantly different density of CD68 and CD163 cells between normal and tumor tissue. Lymph node status, PSA density, and tissue VEGF expression predict recurrence after chemotherapy for high-risk prostate cancer. Additional studies are needed to determine the potential benefit of chemotherapy in the neoadjuvant setting.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Chemotherapy, Adjuvant , Cohort Studies , Docetaxel , Follow-Up Studies , Humans , Male , Middle Aged , Mitoxantrone/administration & dosage , Neoadjuvant Therapy , Prostatectomy , Risk Factors , Taxoids/administration & dosage
17.
J Hepatol ; 64(4): 881-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26639397

ABSTRACT

BACKGROUND & AIMS: The leukocyte composition of tumors is heterogeneous, as is the involvement of each leukocyte subset in promoting or restraining tumorigenesis. This heterogeneity reflects the tissue of origin, tumor stage, and the functional state of leukocyte activation, but its biological roots remain poorly understood. Since tumorigenesis is driven by various genetic events, we assessed the role of driver genes in shaping the profiles and the roles of leukocytes in tumorigenesis. METHODS: Mouse liver tumors were induced by hepatic overexpression of either MYC or the combination of myristoylated AKT and NRAS(V12) oncogenes via hydrodynamic transfection. A comparative, flow cytometry- and histology-based immunophenotyping of liver-infiltrating leukocytes was performed at various stages of liver tumorigenesis. The roles of the most abundant leukocyte subsets in tumorigenesis were addressed by immunodepletion. The contribution of liver injury was assessed by comparing the injury-inducing hydrodynamic transfection model to a model in which MYC is an inducible transgene. RESULTS: Myristoylated AKT and NRAS(V12) promoted a marked recruitment of CD11b(+)Ly6G(hi)Ly6C(int) neutrophils and CD11b(+)Ly6G(-)Ly6C(hi) monocytes to the liver, but their immunodepletion did not alter tumorigenesis. In contrast, despite minimal invasion by monocytes/neutrophils during MYC-driven tumorigenesis, immunodepletion of these cells reduced MYC tumor burden and extended survival. MYC-driven tumor initiation was augmented specifically by Ly6C+ monocytes and their ability to promote liver injury. CONCLUSIONS: Our results demonstrate that leukocyte profiles do not necessarily predict their involvement in tumorigenesis, the functional role of leukocytes can be shaped by oncogenes, and that monocyte-dependent tissue injury selectively cooperates with MYC during tumorigenesis.


Subject(s)
Genes, myc/physiology , Liver Neoplasms, Experimental/etiology , Monocytes/physiology , Animals , Antigens, Ly/analysis , Female , Genes, ras , Mice , Neutrophil Infiltration , Proto-Oncogene Proteins c-akt/genetics , Receptors, Chemokine/analysis
18.
Breast Cancer Res Treat ; 154(3): 495-508, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26590814

ABSTRACT

We previously reported using statins was correlated with improved metastasis-free survival in aggressive breast cancer. The purpose of this study was to examine the effect of statins on metastatic colonization by triple-negative breast cancer (TNBC) cells. TNBC cell lines were treated with simvastatin and then studied for cell cycle progression and proliferation in vitro, and metastasis formation in vivo, following injection of statin-treated cells. Reverse-phase protein assay (RPPA) analysis was performed on statin-treated and control breast cancer cells. RNA interference targeting FOXO3a was used to measure the impact of simvastatin on FOXO3a-expressing cells. The prognostic value of FOXO3a mRNA expression was examined in eight public breast cancer gene expression datasets including 1479 patients. Simvastatin increased G1/S-phase arrest of the cell cycle and inhibited both proliferation and migration of TNBC cells in vitro. In vitro pre-treatment and in vivo treatment with simvastatin reduced metastases. Phosphorylated FOXO3a was downregulated after simvastatin treatment in (RPPA) analysis. Ectopic expression of FOXO3a enhanced mammosphere formation and migratory capacity in vitro. Knockdown of FOXO3a attenuated the effect of simvastatin on mammosphere formation and migration. Analysis of public gene expression data demonstrates FOXO3a mRNA downregulation was independently associated with shorter metastasis-free survival in all breast cancers, as well as in TNBC breast cancers. Simvastatin inhibits in vitro endpoints associated with metastasis through a FOXO3a mechanism and reduced metastasis formation in vivo. FOXO3a expression is prognostic for metastasis formation in patient data. Further investigation of simvastatin as a cancer therapy is warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Transcription Factors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, SCID , Middle Aged , Neoplasm Metastasis , Xenograft Model Antitumor Assays
19.
Trends Immunol ; 33(3): 119-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22277903

ABSTRACT

Of the multiple unique stromal cell types common to solid tumors, tumor-associated macrophages (TAMs) are significant for fostering tumor progression. The protumor properties of TAMs derive from regulation of angiogenic programming, production of soluble mediators that support proliferation, survival and invasion of malignant cells, and direct and indirect suppression of cytotoxic T cell activity. These varied activities are dependent on the polarization state of TAMs that is regulated in part by local concentrations of cytokines and chemokines, as well as varied interactions of TAMs with normal and degraded components of the extracellular matrix. Targeting molecular pathways regulating TAM polarization holds great promise for anticancer therapy.


Subject(s)
Macrophages/immunology , Neoplasms/immunology , Tumor Microenvironment , Animals , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic
20.
J Immunol ; 191(4): 2009-17, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851682

ABSTRACT

Immune cells comprise a substantial proportion of the tumor mass in human nonsmall cell lung cancers (NSCLC), but the precise composition and significance of this infiltration are unclear. In this study, we examined immune complexity of human NSCLC as well as NSCLC developing in CC10-TAg transgenic mice, and revealed that CD4(+) T lymphocytes represent the dominant population of CD45(+) immune cells, and, relative to normal lung tissue, CD4(+)Foxp3(+) regulatory T cells (Tregs) were significantly increased as a proportion of total CD4(+) cells. To assess the functional significance of increased Tregs, we evaluated CD8(+) T cell-deficient/CC10-TAg mice and revealed that CD8(+) T cells significantly controlled tumor growth with antitumor activity that was partially repressed by Tregs. However, whereas treatment with anti-CD25-depleting mAb as monotherapy preferentially depleted Tregs and improved CD8(+) T cell-mediated control of tumor progression during early tumor development, similar monotherapy was ineffective at later stages. Because mice bearing early NSCLC treated with anti-CD25 mAb exhibited increased tumor cell death associated with infiltration by CD8(+) T cells expressing elevated levels of granzyme A, granzyme B, perforin, and IFN-γ, we therefore evaluated carboplatin combination therapy resulting in a significantly extended survival beyond that observed with chemotherapy alone, indicating that Treg depletion in combination with cytotoxic therapy may be beneficial as a treatment strategy for advanced NSCLC.


Subject(s)
Adenocarcinoma/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Carboplatin/administration & dosage , Carboplatin/therapeutic use , Cisplatin/therapeutic use , Cisplatin/toxicity , Cytotoxicity, Immunologic , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Lung Neoplasms/pathology , Lymphocyte Count , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/pathology , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Mutant Strains , Mice, Transgenic , Random Allocation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/pathology , Tumor Escape , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL