Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Rep ; 42(12): 113507, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38041815

ABSTRACT

The expression of pro-lymphangiogenic VEGF-C in primary tumors is associated with sentinel lymph node metastasis in most solid cancer types. However, the impact of VEGF-C on distant organ metastasis remains unclear. Perivascular tumor-associated macrophages (TAMs) play a crucial role in guiding hematogenous spread of cancer cells by establishing metastatic pathways within the tumor microenvironment. This process supports breast cancer cell intravasation and metastatic dissemination. We show here that VEGF-C-expressing TAMs reduce the dissemination of mammary cancer cells to the lungs while concurrently increasing lymph node metastasis. These TAMs express podoplanin and interact with normalized tumor blood vessels expressing VEGFR3. Moreover, clinical data suggest inverse association between VEGF-C-expressing TAMs and breast cancer malignancy. Thus, our study elucidates the paradoxical role of VEGF-C-expressing TAMs in redirecting cancer cells to preferentially disseminate to lymph nodes rather than to lungs, partially achieved by normalizing tumor blood vessels and promoting lymphangiogenesis.


Subject(s)
Breast Neoplasms , Humans , Female , Lymphatic Metastasis , Breast Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Vascular Endothelial Growth Factor C/metabolism , Lymphangiogenesis , Tumor Microenvironment
2.
Nat Rev Cancer ; 23(6): 408-425, 2023 06.
Article in English | MEDLINE | ID: mdl-37142795

ABSTRACT

Cells can rapidly adjust their proteomes in dynamic environments by regulating mRNA translation. There is mounting evidence that dysregulation of mRNA translation supports the survival and adaptation of cancer cells, which has stimulated clinical interest in targeting elements of the translation machinery and, in particular, components of the eukaryotic initiation factor 4F (eIF4F) complex such as eIF4E. However, the effect of targeting mRNA translation on infiltrating immune cells and stromal cells in the tumour microenvironment (TME) has, until recently, remained unexplored. In this Perspective article, we discuss how eIF4F-sensitive mRNA translation controls the phenotypes of key non-transformed cells in the TME, with an emphasis on the underlying therapeutic implications of targeting eIF4F in cancer. As eIF4F-targeting agents are in clinical trials, we propose that a broader understanding of their effect on gene expression in the TME will reveal unappreciated therapeutic vulnerabilities that could be used to improve the efficacy of existing cancer therapies.


Subject(s)
Eukaryotic Initiation Factor-4F , Neoplasms , Humans , Eukaryotic Initiation Factor-4F/genetics , Eukaryotic Initiation Factor-4F/metabolism , Tumor Microenvironment/genetics , Protein Biosynthesis , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Neoplasms/genetics
3.
J Immunother Cancer ; 10(10)2022 10.
Article in English | MEDLINE | ID: mdl-36283735

ABSTRACT

Downregulation of MHC class I (MHCI) molecules on tumor cells is recognized as a resistance mechanism of cancer immunotherapy. Given that MHCI molecules are potent regulators of immune responses, we postulated that the expression of MHCI by tumor cells influences systemic immune responses. Accordingly, mice-bearing MHCI-deficient tumor cells showed reduced tumor-associated extramedullary myelopoiesis in the spleen. Depletion of natural killer (NK) cells abrogated these differences, suggesting an integral role of immune-regulatory NK cells during tumor progression. Cytokine-profiling revealed an upregulation of TNF-α by NK cells in tumors and spleen in mice-bearing MHCI expressing tumors, and inhibition of TNF-α enhanced host myelopoiesis in mice receiving adoptive transfer of tumor-experienced NK cells. Our study highlights a critical role of NK cells beyond its identity as a killer lymphocyte and more importantly, the potential host responses to a localized tumor as determined by its MHCI expression.


Subject(s)
Myelopoiesis , Neoplasms , Mice , Animals , Tumor Necrosis Factor-alpha , Killer Cells, Natural , Histocompatibility Antigens Class I
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649219

ABSTRACT

Infiltration of tumor-promoting immune cells is a strong driver of tumor progression. Especially the accumulation of macrophages in the tumor microenvironment is known to facilitate tumor growth and to correlate with poor prognosis in many tumor types. TAp73, a member of the p53/p63/p73 family, acts as a tumor suppressor and has been shown to suppress tumor angiogenesis. However, what role TAp73 has in regulating immune cell infiltration is unknown. Here, we report that low levels of TAp73 correlate with an increased NF-κB-regulated inflammatory signature in breast cancer. Furthermore, we show that loss of TAp73 results in NF-κB hyperactivation and secretion of Ccl2, a known NF-κB target and chemoattractant for monocytes and macrophages. Importantly, TAp73-deficient tumors display an increased accumulation of protumoral macrophages that express the mannose receptor (CD206) and scavenger receptor A (CD204) compared to controls. The relevance of TAp73 expression in human breast carcinoma was further accentuated by revealing that TAp73 expression correlates negatively with the accumulation of protumoral CD163+ macrophages in breast cancer patient samples. Taken together, our findings suggest that TAp73 regulates macrophage accumulation and phenotype in breast cancer through inhibition of the NF-κB pathway.


Subject(s)
Breast Neoplasms/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Tumor Protein p73/immunology , Tumor-Associated Macrophages/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Breast Neoplasms/pathology , Chemokine CCL2/immunology , Female , Humans , Membrane Glycoproteins/immunology , Mice , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Scavenger Receptors, Class A/immunology , Tumor-Associated Macrophages/pathology
5.
Gynecol Oncol ; 159(3): 860-868, 2020 12.
Article in English | MEDLINE | ID: mdl-33032823

ABSTRACT

OBJECTIVE: Pre-clinical studies have identified marker- and tumor compartment-defined functionally distinct macrophage subsets. Our study analyzes marker-defined macrophage subsets in different tumor compartments of high-grade serous ovarian cancer (HGSC). METHODS: A discovery cohort (N = 113) was subjected to immunohistochemistry (IHC) analyses. CD68-positivity was confirmed for CD11c-, CD80- and CD163-positive cells. Subset-marker-positive cells were scored in the total tumor and in four tumor compartments. Correlation analyses investigated co-expression of subsets, relationship to CD8+ cells and survival associations. A validation cohort (N = 121) was used to confirm selected findings from the discovery cohort. RESULTS: CD163-positve cells was the most abundant subtype in all compartments. CD11c and CD163 subsets were strongly correlated with each other in stroma and epithelial areas, whereas CD80 and CD163 were correlated in epithelial areas. CD80 and CD11c in perivascular areas showed low correlations. Strong associations were detected between CD8 and CD80 in the tumor epithelium-dominated areas, and between CD8 and CD11c in stroma areas. High stromal CD11c density was associated with a longer median overall survival in the discovery cohort (HR 0.39; CI 95%, 0.23-0.68; p = 0.001) and in the validation cohort (HR 0.46; CI 95%, 0.22-0.93; p = 0.03). CONCLUSIONS: Our study supports the existence of clinically relevant marker- and localization defined macrophage subsets in HGSC, which are independently regulated. Moreover, it suggests stromal CD11c as a novel prognostic marker in HGSC.


Subject(s)
Biomarkers, Tumor/metabolism , CD11c Antigen/metabolism , Carcinoma, Ovarian Epithelial/mortality , Ovarian Neoplasms/mortality , Tumor-Associated Macrophages/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovary/immunology , Ovary/pathology , Prognosis , Retrospective Studies , Sweden , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism
6.
Proc Natl Acad Sci U S A ; 117(44): 27556-27565, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33077599

ABSTRACT

Tumor-associated macrophages (TAMs) continuously fine tune their immune modulatory properties, but how gene expression programs coordinate this immune cell plasticity is largely unknown. Selective mRNA translation, controlled by MNK1/MNK2 and mTOR pathways impinging on eIF4E, facilitates reshaping of proteomes without changes in abundance of corresponding mRNAs. Using polysome profiling developed for small samples we show that, during tumor growth, gene expression in TAMs is predominately modulated via mRNA-selective changes in translational efficiencies. These alterations in gene expression paralleled accumulation of antiinflammatory macrophages with augmented phosphorylation of eIF4E, a target of the MNK1 and MNK2 kinases, known to selectively modulate mRNA translation. Furthermore, suppression of the MNK2, but not the mTOR signaling pathway, reprogrammed antiinflammatory macrophages toward a proinflammatory phenotype with the ability to activate CD8+ T cells. Thus, selective changes of mRNA translation depending on MNK2 signaling represents a key node regulating macrophage antiinflammatory functions.


Subject(s)
Macrophages/immunology , Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , Animals , Coculture Techniques , Disease Models, Animal , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Female , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Humans , MCF-7 Cells , Macrophages/metabolism , Mice , Mice, Transgenic , Naphthyridines/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/genetics , Phosphorylation/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Escape/genetics
7.
Cancers (Basel) ; 11(10)2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31581535

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is an oncogene and multifaceted transcription factor involved in multiple cellular functions. Its role in modifying anti-tumor immunity has been recently recognized. In this study, the biologic effects of STAT3 on immune checkpoint expression and anti-tumor responses were investigated in breast cancer (BC). A transcriptional signature of phosphorylated STAT3 was positively correlated with PD-L1 expression in two independent cohorts of early BC. Pharmacologic inhibition and gene silencing of STAT3 led to decreased Programmed Death Ligand 1 (PD-L1) expression levels in vitro, and resulted as well in reduction of tumor growth and decreased metastatic dissemination in a mammary carcinoma mouse model. The hampering of tumor progression was correlated to an anti-tumoral macrophage phenotype and accumulation of natural-killer cells, but also in reduced accrual of cytotoxic lymphocytes. In human BC, pro-tumoral macrophages correlated to PD-L1 expression, proliferation status and higher grade of malignancy, indicating a subset of patients with immunosuppressive properties. In conclusion, this study provides evidence for STAT3-mediated regulation of PD-L1 and modulation of immune microenvironment in BC.

8.
Clin Cancer Res ; 25(12): 3702-3717, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30850359

ABSTRACT

PURPOSE: Fibroblasts expressing the orphan chemokine CXCL14 have been previously shown to associate with poor breast cancer prognosis and promote cancer growth. This study explores the mechanism underlying the poor survival associations of stromal CXCL14. EXPERIMENTAL DESIGN: Tumor cell epithelial-to-mesenchymal transition (EMT), invasion, and metastasis were studied in in vitro and in vivo models together with fibroblasts overexpressing CXCL14. An approach for CXCL14 receptor identification included loss-of-function studies followed by molecular and functional endpoints. The clinical relevance was further explored in publicly available gene expression datasets. RESULTS: CXCL14 fibroblasts stimulated breast cancer EMT, migration, and invasion in breast cancer cells and in a xenograft model. Furthermore, tumor cells primed by CXCL14 fibroblasts displayed enhanced lung colonization after tail-vein injection. By loss-of function experiments, the atypical G-protein-coupled receptor ACKR2 was identified to mediate CXCL14-stimulated responses. Downregulation of ACKR2, or CXCL14-induced NOS1, attenuated the pro-EMT and migratory capacity. CXCL14/ACKR2 expression correlated with EMT and survival in gene expression datasets. CONCLUSIONS: Collectively, the findings imply an autocrine fibroblast CXCL14/ACKR2 pathway as a clinically relevant stimulator of EMT, tumor cell invasion, and metastasis. The study also identifies ACKR2 as a novel mediator for CXCL14 function and thereby defines a pathway with drug target potential.See related commentary by Zhang et al., p. 3476.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Chemokines, CXC/genetics , Fibroblasts , Gene Expression Regulation, Neoplastic , Humans
9.
Am J Respir Crit Care Med ; 200(3): 348-358, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30742544

ABSTRACT

Rationale: Chronic obstructive pulmonary disease is an independent risk factor for lung cancer, but the underlying molecular mechanisms are unknown. We hypothesized that lung stromal cells activate pathological gene expression programs that support oncogenesis.Objectives: To identify molecular mechanisms operating in the lung stroma that support the development of lung cancer.Methods: The study included subjects with and without lung cancer across a spectrum of lung-function values. We conducted a multiomics analysis of nonmalignant lung tissue to quantify the transcriptome, translatome, and proteome.Measurements and Main Results: Cancer-associated gene expression changes predominantly manifested as alterations in the efficiency of mRNA translation modulating protein levels in the absence of corresponding changes in mRNA levels. The molecular mechanisms that drove these cancer-associated translation programs differed based on lung function. In subjects with normal to mildly impaired lung function, the mammalian target of rapamycin (mTOR) pathway served as an upstream driver, whereas in subjects with severe airflow obstruction, pathways downstream of pathological extracellular matrix emerged. Consistent with a role during cancer initiation, both the mTOR and extracellular matrix gene expression programs paralleled the activation of previously identified procancer secretomes. Furthermore, an in situ examination of lung tissue showed that stromal fibroblasts expressed cancer-associated proteins from two procancer secretomes: one that included IL-6 (in cases of mild or no airflow obstruction), and one that included BMP1 (in cases of severe airflow obstruction).Conclusions: Two distinct stromal gene expression programs that promote cancer initiation are activated in patients with lung cancer depending on lung function. Our work has implications both for screening strategies and for personalized approaches to cancer treatment.


Subject(s)
Lung Neoplasms/etiology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , Stromal Cells/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Forced Expiratory Volume , Humans , Male , Middle Aged , Proteome , Pulmonary Disease, Chronic Obstructive/pathology , Transcriptome
10.
iScience ; 9: 71-83, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30384135

ABSTRACT

High-grade gliomas (HGGs) are the most aggressive and invasive primary brain tumors. The platelet-derived growth factor (PDGF) signaling pathway drives HGG progression, and enhanced expression of PDGF receptors (PDGFRs) is a well-established aberration in a subset of glioblastomas (GBMs). PDGFRA is expressed in glioma cells, whereas PDGFRB is mostly restricted to the glioma-associated stroma. Here we show that the spatial location of TAMMs correlates with the expansion of a subset of tumor cells that have acquired expression of PDGFRB in both mouse and human low-grade glioma and HCGs. Furthermore, M2-polarized microglia but not bone marrow (BM)-derived macrophages (BMDMs) induced PDGFRB expression in glioma cells and stimulated their migratory capacity. These findings illustrate a heterotypic cross-talk between microglia and glioma cells that may enhance the migratory and invasive capacity of the latter by inducing PDGFRB.

11.
Cancer Immunol Res ; 6(11): 1417-1425, 2018 11.
Article in English | MEDLINE | ID: mdl-30143536

ABSTRACT

Metastatic breast cancer is a fatal disease that responds poorly to treatment. Cancer vaccines targeting antigens expressed by metastatic breast cancer cells and cancer stem cells could function as anticancer therapies. Cripto-1 is an oncofetal protein overexpressed in invasive breast cancer and cancer-initiating cells. In this study, we explored the potential of a Cripto-1-encoding DNA vaccine to target breast cancer in preclinical mouse models. BALB/c mice and BALB-neuT mice were treated with a DNA vaccine encoding mouse Cripto-1 (mCr-1). BALB/c mice were challenged with murine breast cancer 4T1 cells or TUBO spheres; BALB-neuT mice spontaneously developed breast cancer. Tumor growth was followed in all mouse models and lung metastases were evaluated. In vitro assays were performed to identify the immune response elicited by vaccination. Vaccination against mCr-1 reduced primary tumor growth in the 4T1 metastatic breast cancer model and reduced lung metastatic burden. In BALB-neuT mice, because the primary tumors are Cripto-1 negative, vaccination against mCr-1 did not affect primary tumors but did reduce lung metastatic burden. Spheroid-cultured TUBO cells, derived from a BALB/neuT primary tumor, develop a cancer stem cell-like phenotype and express mCr-1. We observed reduced tumor growth in vaccinated mice after challenge with TUBO spheres. Our data indicate that vaccination against Cripto-1 results in a protective immune response against mCr-1 expressing and metastasizing cells. Targeting Cripto-1 by vaccination holds promise as an immunotherapy for treatment of metastatic breast cancer. Cancer Immunol Res; 6(11); 1417-25. ©2018 AACR.


Subject(s)
Cancer Vaccines/pharmacology , Epidermal Growth Factor/genetics , Mammary Neoplasms, Experimental/therapy , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Vaccines, DNA/pharmacology , Animals , Cell Line, Tumor , Epidermal Growth Factor/immunology , Female , Immunity, Humoral , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Membrane Glycoproteins/immunology , Mice, Inbred BALB C , Neoplasm Proteins/immunology , Neoplastic Stem Cells/immunology
12.
Oncoimmunology ; 6(8): e1338238, 2017.
Article in English | MEDLINE | ID: mdl-28920001

ABSTRACT

Regulatory T cells (Treg) suppress anti-tumor immune responses and their infiltration in the tumor microenvironment is associated with inferior prognosis in cancer patients. Thus, in order to enhance anti-tumor immune responses, selective depletion of Treg is highly desired. We found that treatment with zoledronic acid (ZA) resulted in a selective decrease in the frequency of Treg that was associated with a significant increase in proliferation of T cells and natural killer (NK) cells in peripheral blood of patients with metastatic cancer. In vitro, genome-wide transcriptomic analysis revealed alterations in calcium signaling pathways in Treg following treatment with ZA. Furthermore, co-localization of the nuclear factor of activated T cells (NFAT) and forkhead box P3 (FOXP3) was significantly reduced in Treg upon ZA-treatment. Consequently, reduced expression levels of CD25, STAT5 and TGFß were observed. Functionally, ZA-treated Treg had reduced capacity to suppress T and NK cell proliferation and anti-tumor responses compared with untreated Treg in vitro. Treatment with ZA to selectively inhibit essential signaling pathways in Treg resulting in reduced capacity to suppress effector T and NK cell responses represents a novel approach to inhibit Treg activity in patients with cancer.

13.
Cell Rep ; 15(9): 2000-11, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27210762

ABSTRACT

Tumors are composed of multiple cell types besides the tumor cells themselves, including innate immune cells such as macrophages. Tumor-associated macrophages (TAMs) are a heterogeneous population of myeloid cells present in the tumor microenvironment (TME). Here, they contribute to immunosuppression, enabling the establishment and persistence of solid tumors as well as metastatic dissemination. We have found that the pattern recognition scavenger receptor MARCO defines a subtype of suppressive TAMs and is linked to clinical outcome. An anti-MARCO monoclonal antibody was developed, which induces anti-tumor activity in breast and colon carcinoma, as well as in melanoma models through reprogramming TAM populations to a pro-inflammatory phenotype and increasing tumor immunogenicity. This anti-tumor activity is dependent on the inhibitory Fc-receptor, FcγRIIB, and also enhances the efficacy of checkpoint therapy. These results demonstrate that immunotherapies using antibodies designed to modify myeloid cells of the TME represent a promising mode of cancer treatment.


Subject(s)
Antibodies, Neoplasm/pharmacology , Disease Progression , Macrophages/metabolism , Neoplasms/pathology , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cytokines/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Immunosuppression Therapy , Immunotherapy , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/therapy , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Cancer Res ; 76(11): 3166-78, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27197153

ABSTRACT

Accumulation of tumor-associated macrophages (TAM) correlates with malignant progression, immune suppression, and poor prognosis. In this study, we defined a critical role for the cell-surface guidance molecule SEMA3A in differential proliferative control of TAMs. Tumor cell-derived SEMA3A restricted the proliferation of protumoral M2 macrophages but increased the proliferation of antitumoral M1, acting through the SEMA3A receptor neuropilin 1. Expansion of M1 macrophages in vivo enhanced the recruitment and activation of natural killer (NK) cells and cytotoxic CD8(+) T cells to tumors, inhibiting their growth. In human breast cancer specimens, we found that immunohistochemical levels of SEMA3A correlated with the expression of genes characteristic of M1 macrophages, CD8(+) T cells, and NK cells, while inversely correlating with established characters of malignancy. In summary, our results illuminate a mechanism whereby the TAM phenotype is controlled and identify the cell-surface molecule SEMA3A as a candidate for therapeutic targeting. Cancer Res; 76(11); 3166-78. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Killer Cells, Natural/pathology , Macrophages/pathology , Semaphorin-3A/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Disease Progression , Female , Humans , Immunoenzyme Techniques , Killer Cells, Natural/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Neoplasm Grading , Neuropilin-1/genetics , Neuropilin-1/metabolism , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Semaphorin-3A/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Science ; 347(6226): 1138-42, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25700174

ABSTRACT

The mammalian cerebral cortex supports cognitive functions such as sensorimotor integration, memory, and social behaviors. Normal brain function relies on a diverse set of differentiated cell types, including neurons, glia, and vasculature. Here, we have used large-scale single-cell RNA sequencing (RNA-seq) to classify cells in the mouse somatosensory cortex and hippocampal CA1 region. We found 47 molecularly distinct subclasses, comprising all known major cell types in the cortex. We identified numerous marker genes, which allowed alignment with known cell types, morphology, and location. We found a layer I interneuron expressing Pax6 and a distinct postmitotic oligodendrocyte subclass marked by Itpr2. Across the diversity of cortical cell types, transcription factors formed a complex, layered regulatory code, suggesting a mechanism for the maintenance of adult cell type identity.


Subject(s)
CA1 Region, Hippocampal/cytology , Interneurons/classification , Oligodendroglia/classification , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Somatosensory Cortex/cytology , Animals , CA1 Region, Hippocampal/metabolism , Eye Proteins/genetics , Gene Expression , Genetic Markers , Homeodomain Proteins/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Interneurons/cytology , Interneurons/metabolism , Mice , Oligodendroglia/cytology , Oligodendroglia/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Phylogeny , Repressor Proteins/genetics , Somatosensory Cortex/metabolism , Transcription Factors/classification , Transcription Factors/genetics , Transcriptome
17.
Eur J Immunol ; 43(1): 249-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22996291

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that are able to directly kill tumor cells through different mechanisms including ligation of TNF-related apoptosis-inducing ligand (TRAIL) receptors. Zoledronic acid (ZA) is a bisphosphonate known to upregulate the expression of TRAIL on human γδ T cells. Here, we investigated whether exposure to ZA would upregulate TRAIL expression on human NK cells and augment their cytotoxicity against tumor cells. When cocultured with monocytes, treatment with ZA and IL-2 resulted in a significant upregulation of TRAIL expression on human NK cells (p = 0.002). Consequently, ZA-primed NK cells were significantly more cytotoxic against TRAIL sensitive tumor cells (p < 0.0001). In the presence of ZA and IL-2, monocytes produced high levels of IFN-γ; when cultured in the presence of neutralizing antibodies to IFN-γ, TRAIL expression and TRAIL-mediated cytotoxicity of NK cells were significantly reduced. Furthermore, in tumor-bearing SCID/Beige mice, a significant delayed tumor progression and prolonged survival was observed after infusion of ZA-primed NK cells compared with that observed in mice infused with unprimed NK cells. These findings represent a novel approach to potentiate TRAIL-mediated apoptosis by adoptively infused NK cells that could improve the outcome in patients with cancer.


Subject(s)
Cytotoxicity, Immunologic , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Monocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cells, Cultured , Coculture Techniques , Diphosphonates/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Imidazoles/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Lymphocyte Activation/drug effects , Mice , Mice, SCID , Neoplasms/immunology , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays , Zoledronic Acid
18.
Cancer Res ; 72(8): 1953-63, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22374984

ABSTRACT

Histidine-rich glycoprotein (HRG) is a 75-kDa heparin-binding plasma protein implicated in the regulation of tumor growth and vascularization. In this study, we show that hrg(-/-) mice challenged with fibrosarcoma or pancreatic carcinoma grow larger tumors with increased metastatic properties. Compared with wild-type mice, fibrosarcomas in hrg(-/-) mice were more hypoxic, necrotic, and less perfused, indicating enhanced vessel abnormalization. HRG deficiency was associated with a suppressed antitumor immune response, with both increased infiltration of M2 marker-expressing macrophages and decreased infiltration of dendritic cells and cytotoxic T cells. Analysis of transcript expression in tumor-associated as well as peritoneal macrophages from hrg(-/-) mice revealed an increased expression of genes associated with a proangiogenic and immunoinhibitory phenotype. In accordance, expression arrays conducted on HRG-treated peritoneal macrophages showed induction of genes involved in extracellular matrix biology and immune responsiveness. In conclusion, our findings show that macrophages are a direct target of HRG. HRG loss influences macrophage gene regulation, leading to excessive stimulation of tumor angiogenesis, suppression of tumor immune response, and increased tumor growth and metastatic spread.


Subject(s)
Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Proteins/metabolism , Tumor Escape/physiology , Animals , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Proteins/genetics , Proteins/immunology , Tumor Microenvironment/physiology
19.
Arterioscler Thromb Vasc Biol ; 31(4): 741-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205984

ABSTRACT

OBJECTIVE: The role of semaphorins in tumor progression is still poorly understood. In this study, we aimed at elucidating the regulatory role of semaphorin 3A (SEMA3A) in primary tumor growth and metastatic dissemination. METHODS AND RESULTS: We used 3 different experimental approaches in mouse tumor models: (1) overexpression of SEMA3A in tumor cells, (2) systemic expression of SEMA3A following liver gene transfer in mice, and (3) tumor-targeted release of SEMA3A using gene modified Tie2-expressing monocytes as delivery vehicles. In each of these experimental settings, SEMA3A efficiently inhibited tumor growth by inhibiting vessel function and increasing tumor hypoxia and necrosis, without promoting metastasis. We further show that the expression of the receptor neuropilin-1 in tumor cells is required for SEMA3A-dependent inhibition of tumor cell migration in vitro and metastatic spreading in vivo. CONCLUSIONS: In sum, both systemic and tumor-targeted delivery of SEMA3A inhibits tumor angiogenesis and tumor growth in multiple mouse models; moreover, SEMA3A inhibits the metastatic spreading from primary tumors. These data support the rationale for further investigation of SEMA3A as an anticancer molecule.


Subject(s)
Breast Neoplasms/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Lung Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Semaphorin-3A/metabolism , Stem Cells/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Necrosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neuropilin-1/metabolism , Paracrine Communication , RNA Interference , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Recombinant Fusion Proteins/metabolism , Semaphorin-3A/genetics , Signal Transduction , Stromal Cells/metabolism , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
20.
Cancer Cell ; 19(1): 31-44, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21215706

ABSTRACT

Polarization of tumor-associated macrophages (TAMs) to a proangiogenic/immune-suppressive (M2-like) phenotype and abnormal, hypoperfused vessels are hallmarks of malignancy, but their molecular basis and interrelationship remains enigmatic. We report that the host-produced histidine-rich glycoprotein (HRG) inhibits tumor growth and metastasis, while improving chemotherapy. By skewing TAM polarization away from the M2- to a tumor-inhibiting M1-like phenotype, HRG promotes antitumor immune responses and vessel normalization, effects known to decrease tumor growth and metastasis and to enhance chemotherapy. Skewing of TAM polarization by HRG relies substantially on downregulation of placental growth factor (PlGF). Besides unveiling an important role for TAM polarization in tumor vessel abnormalization, and its regulation by HRG/PlGF, these findings offer therapeutic opportunities for anticancer and antiangiogenic treatment.


Subject(s)
Down-Regulation/genetics , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Pregnancy Proteins/metabolism , Proteins/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotactic Factors/metabolism , Clodronic Acid/pharmacology , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Hypoxia/genetics , Hypoxia/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/pathology , Microvessels/ultrastructure , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/immunology , Proteins/genetics , Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...