Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Adv Biol (Weinh) ; 7(12): e2300168, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37615259

ABSTRACT

Hyaluronan (HA) is one of the major components of the extracellular matrix in tumor tissue. Recent reports have made it clear that the balance of HA synthesis and degradation is critical for tumor progression. HA is synthesized on the cytoplasmic surface of the plasma membrane by hyaluronan synthases (HAS) and extruded into the extracellular space. Excessive HA production in cancer is associated with enhanced HA degradation in the tumor microenvironment, leading to the accumulation of HA fragments with small molecular weight. These perturbations in both HA synthesis and degradation may play important roles in tumor progression. Recently, it has become increasingly clear that small HA fragments can induce a variety of biological events, such as angiogenesis, cancer-promoting inflammation, and tumor-associated immune suppression. Progression of urologic malignancies, particularly of prostate and bladder cancers, as well as of certain types of kidney cancer show markedly perturbed metabolism of tumor-associated HA. This review highlights the recent research findings regarding HA metabolism in tumor microenvironments with a special focus on urologic cancers. It also will discuss the potential implications of these findings for the development of novel therapeutic interventions for the treatment of prostate, bladder, and kidney cancers.


Subject(s)
Hyaluronic Acid , Urologic Neoplasms , Male , Humans , Hyaluronic Acid/metabolism , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Urologic Neoplasms/metabolism , Inflammation/metabolism , Extracellular Matrix/metabolism , Tumor Microenvironment
2.
Methods Mol Biol ; 2684: 167-175, 2023.
Article in English | MEDLINE | ID: mdl-37410234

ABSTRACT

Hyaluronan is a major component of the extracellular matrix in both normal and tumor tissue. Many solid cancers, including bladder cancer, are characterized by deregulated hyaluronan metabolism. It is postulated that the deregulated metabolism in cancer tissue is characterized by elevated hyaluronan synthesis and degradation. This results in the accumulation of small hyaluronan fragments in the tumor microenvironment which promotes cancer-related inflammation, stimulates tumor cell proliferation and angiogenesis, and contributes to immune-associated immune suppression. For a better understanding of the complex mechanisms of hyaluronan metabolism in cancer, it has been proposed to use precision-cut tissue slice cultures prepared using freshly excised cancer tissue. Here we describe the protocol for establishing tissue slice cultures and analysis of tumor-associated hyaluronan in human urothelial carcinoma.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Hyaluronic Acid/metabolism , Extracellular Matrix/metabolism , Immunologic Tests , Hyaluronan Receptors/metabolism , Tumor Microenvironment
3.
Front Oncol ; 13: 1208196, 2023.
Article in English | MEDLINE | ID: mdl-37207141
4.
Front Immunol ; 13: 971278, 2022.
Article in English | MEDLINE | ID: mdl-36238286

ABSTRACT

Hyaluronan (HA) is known to be a prominent component of the extracellular matrix in tumors, and many solid cancers are characterized by aberrant HA metabolism resulting in increased production in tumor tissue. HA has been implicated in regulating a variety of cellular functions in tumor cells and tumor-associated stromal cells, suggesting that altered HA metabolism can influence tumor growth and malignancy at multiple levels. Importantly, increased HA production in cancer is associated with enhanced HA degradation due to high levels of expression and activity of hyaluronidases (Hyal). Understanding the complex molecular and cellular mechanisms involved in abnormal HA metabolism and catabolism in solid cancers could have important implications for the design of future cancer therapeutic approaches. It appears that extensive crosstalk between immune cells and HA-enriched stroma contributes to tumor growth and progression in several ways. Specifically, the interaction of tumor-recruited Hyal2-expressing myeloid-derived suppressor cells (MDSCs) of bone marrow origin with HA-producing cancer-associated fibroblasts and epithelial tumor cells results in enhanced HA degradation and accumulation of small pro-inflammatory HA fragments, which further drives cancer-related inflammation. In addition, hyaluronan-enriched stroma supports the transition of tumor-recruited Hyal2+MDSCs to the PD-L1+ tumor-associated macrophages leading to the formation of an immunosuppressive and tolerogenic tumor microenvironment. In this review, we aim to discuss the contribution of tumor-associated HA to cancer inflammation, angiogenesis, and tumor-associated immune suppression. We also highlight the recent findings related to the enhanced HA degradation in the tumor microenvironment.


Subject(s)
Neoplasms , Tumor Microenvironment , B7-H1 Antigen , Humans , Hyaluronic Acid/metabolism , Inflammation , Neoplasms/pathology
6.
J Immunol ; 208(12): 2829-2836, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35589125

ABSTRACT

Expression of the transmembrane protein PD-L1 is frequently upregulated in cancer. Because PD-L1-expressing cells can induce apoptosis or anergy of T lymphocytes through binding to the PD1 receptor, the PD-L1-mediated inhibition of activated PD1+ T cells is considered a major pathway for tumor immune escape. However, the mechanisms that regulate the expression of PD-L1 in the tumor microenvironment are not fully understood. Analysis of organotypic tumor tissue slice cultures, obtained from mice with implanted syngeneic tumors (MBT2 bladder tumors in C3H mice, Renca kidney, and CT26 colon tumors in BALB/c mice), as well as from patients with cancer, revealed that tumor-associated hyaluronan (HA) supports the development of immunosuppressive PD-L1+ macrophages. Using genetically modified tumor cells, we identified epithelial tumor cells and cancer-associated mesenchymal fibroblast-like cells as a major source of HA in the tumor microenvironment. These HA-producing tumor cells, and particularly the vimentin-positive fibroblast-like cells of bone marrow origin, directly interact with tumor-recruited myeloid cells to form large stromal congregates/clusters that are highly enriched for both HA and PD-L1. Furthermore, similar cell clusters composed of HA-producing fibroblast-like cells and PD-L1+ macrophages were detected in tumor-draining, but not in distant, lymph nodes. Collectively, our findings indicate that the formation of multiple large HA-enriched stromal clusters that support the development of PD-L1-expressing APCs in the tumor microenvironment and draining lymph nodes could contribute to the immune escape and resistance to immunotherapy in cancer.


Subject(s)
B7-H1 Antigen , Urinary Bladder Neoplasms , Animals , Cell Line, Tumor , Hyaluronic Acid/metabolism , Lymph Nodes , Macrophages , Mice , Mice, Inbred C3H , Tumor Microenvironment
7.
J Kidney Cancer VHL ; 9(2): 1-6, 2022.
Article in English | MEDLINE | ID: mdl-35528727

ABSTRACT

Renal cell carcinoma (RCC) patients frequently have increased number of immunosuppressive myeloid cells in circulation. High number of myeloid-derived suppressor cells (MDSCs) in the blood are associated with immune suppression as well as with cancer-related inflammation which drives the mobilization of myeloid cells to tumor tissue. Here, we show that peripheral blood from a previously untreated RCC patient has increased the number of monocytic CD33+CD11b+ MDSCs, which also co-expressed PD-L1 and membrane-bound enzyme hyaluronidase 2 (Hyal2). PD-L1 expression is associated with immune suppression, whereas expression of Hyal2 is associated with inflammation, because Hyal2+ myeloid cells can degrade the extracellular hyaluronan (HA), leading to the accumulation of pro-inflammatory HA fragments with low molecular weight. These findings implicate the potential involvement of monocytic MDSCs in both tumor-associated immune suppression and cancer-related inflammation. Analysis of organotypic tumor-tissue slice cultures prepared from cancer tissue of the same patient revealed the significant presence of PD-L1+ HLA-DR+ macrophage-like or dendritic cell-like antigen-presenting cells in tumor stroma. Interestingly, stroma-associated PD-L1+ cells frequently have intracellular hyaluronan. Collectively, data presented in this study suggest that the interplay between tumor-recruited myeloid cells and stromal HA may contribute to the inflammation and immune tolerance in kidney cancer.

9.
Cancer Res ; 81(10): 2584-2585, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33999840

ABSTRACT

In this issue of Cancer Research, Zhou and colleagues investigate the role of acute kidney injury (AKI) and AKI-associated systemic inflammation in the development of kidney cancer. They demonstrate a positive association between the formation of clear-cell renal cell carcinoma and AKI induced by ischemia-reperfusion injury in genetically modified mice. In parallel with the emergence of kidney tumors, mice with ischemic injury develop systemic inflammation associated with tissue infiltration by neutrophils and fibroblasts and upregulated expression of several inflammatory factors, with CXCL1 displaying the highest levels of upregulation. Accordingly, blockade of CXCL1-mediated signaling inhibited the emergence of kidney tumors in mice subjected to ischemic kidney injury. The study provides evidence for a new experimental approach to prevent the formation of clear-cell renal cell carcinoma and reduce kidney cancer incidence through modulation of the AKI-induced inflammatory response using inhibitors of CXC/CXCR2 axis. As the incidence of kidney cancer continues to increase, new treatment strategies for this devastating disease are urgently needed. Zhou and colleagues provide preclinical proof of concept for a new therapeutic strategy and address an unmet need for this difficult to prevent and treat cancer disease.See related article by Zhou et al., p. 2690.


Subject(s)
Acute Kidney Injury , Carcinoma, Renal Cell , Kidney Neoplasms , Acute Kidney Injury/etiology , Animals , Epithelial Cells , Inflammation , Kidney , Mice
10.
Cancer Res ; 81(3): 648-657, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33239427

ABSTRACT

The increased presence of myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) in tumor tissue has been extensively reported. However, their role in the regulation of hyaluronan (HA) metabolism in the tumor microenvironment has not been established. Here we describe a novel function of tumor-associated myeloid cells related to the enhanced breakdown of extracellular HA in human bladder cancer tissue, leading to the accumulation of small HA fragments with molecular weight (MW) <20 kDa. Increased fragmentation of extracellular HA and accumulation of low molecular weight HA (LMW-HA) in tumor tissue was associated with elevated production of multiple inflammatory cytokines, chemokines, and angiogenic factors. The fragmentation of HA by myeloid cells was mediated by the membrane-bound enzyme hyaluronidase 2 (Hyal2). Increased numbers of Hyal2+CD11b+ myeloid cells were detected in the tumor tissue as well as in the peripheral blood of patients with bladder cancer. Coexpression of CD33 suggested that these cells belong to monocytic myeloid-derived suppressor cells. The HA-degrading function of Hyal2-expressing MDSCs could be enhanced by exposure to tumor-conditioned medium, and IL1ß was identified as one of the factors involved in the stimulation of Hyal2 activity. CD44-mediated signaling played an important role in the regulation of HA-degrading activity of Hyal2-expressing myeloid cells, as the engagement of CD44 receptor with specific mAb triggered translocation of Hyal2 enzyme to the cellular surface and stimulated secretion of IL1ß. Taken together, this work identifies Hyal2-expressing tumor-associated myeloid cells as key players in the accumulation of LMW-HA in the tumor microenvironment and cancer-related inflammation and angiogenesis. SIGNIFICANCE: This study identifies Hyal2-expressing tumor-associated myeloid cells of monocyte-macrophage lineage as contributors to hyaluronan degradation in bladder cancer tissue, leading to accumulation of inflammatory and proangiogenic low molecular weight hyaluronan fragments.


Subject(s)
Cell Adhesion Molecules/metabolism , Hyaluronoglucosaminidase/metabolism , Inflammation/metabolism , Myeloid Cells/metabolism , Urinary Bladder Neoplasms/metabolism , Cell Line, Tumor , Chemokines/metabolism , Cytokines/metabolism , GPI-Linked Proteins/metabolism , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Inflammation/pathology , Molecular Weight , Myeloid Cells/pathology , Tumor Microenvironment , Urinary Bladder Neoplasms/pathology
11.
Cancer Immunol Immunother ; 69(1): 3-14, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31811337

ABSTRACT

With the introduction of multiple new agents, the role of immunotherapy is rapidly expanding across all malignancies. Bladder cancer is known to be immunogenic and is responsive to immunotherapy including intravesical BCG and immune checkpoint inhibitors. Multiple trials have addressed the role of checkpoint inhibitors in advanced bladder cancer, including atezolizumab, avelumab, durvalumab, nivolumab and pembrolizumab (all targeting the PD1/PD-L1 pathway). While these trials have demonstrated promising results and improvements over existing therapies, less than half of patients with advanced disease demonstrate clinical benefit from checkpoint inhibitor therapy. Recent breakthroughs in cancer biology and immunology have led to an improved understanding of the influence of the tumor microenvironment on the host's immune system. It appears that tumors promote the formation of highly immunosuppressive microenvironments preventing generation of effective anti-tumor immune response through multiple mechanisms. Therefore, reconditioning of the tumor microenvironment and restoration of the competent immune response is essential for achieving optimal efficacy of cancer immunotherapy. In this review, we aim to discuss the major mechanisms of immune evasion in bladder cancer and highlight novel pathways and molecular targets that may help to attenuate tumor-induced immune tolerance, overcome resistance to immunotherapy and improve clinical outcomes.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Escape/immunology , Urinary Bladder Neoplasms/immunology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Humans , Molecular Targeted Therapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology , Treatment Outcome , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/drug therapy
12.
Front Immunol ; 9: 1863, 2018.
Article in English | MEDLINE | ID: mdl-30186283

ABSTRACT

Purpose: A number of hyperoxaluric states have been associated with calcium oxalate (CaOx) deposits in the kidneys. In animal models of stone disease, these crystals interact with circulating monocytes that have migrated into the kidney as part of innate immunity. Similarly, macrophages surround CaOx crystals in kidneys of patients excreting high levels of oxalate. We investigate the effect of this exposure and subsequent human immunological response in vitro. Materials and methods: Primary human monocytes were collected from healthy donors and exposed to CaOx, potassium oxalate, and zinc oxalate (ZnOx). Cytokine production was measured with a multiplex ELISA. Quantitative reverse transcription-polymerase chain reaction was done to validate the mRNA profile expression. M1 macrophage phenotype was confirmed with immunofluorescence microscopy. Results: Both primary monocytes and THP-1 cells, a human monocytic cell line, respond strongly to CaOx crystals in a dose-dependent manner producing TNF-α, IL-1ß, IL-8, and IL-10 transcripts. Exposure to CaOx followed by 1 h with LPS had an additive effect for cytokine production compared to LPS alone, however, LPS followed by CaOx led to significant decrease in cytokine production. Supernatants taken from monocytes were previously exposed to CaOx crystals enhance M2 macrophage crystal phagocytosis. CaOx, but not potassium or ZnOx, promotes monocyte differentiation into inflammatory M1-like macrophages. Conclusion: In our in vitro experiment, human monocytes were activated by CaOx and produced inflammatory cytokines. Monocytes recognized CaOx crystals through a specific mechanism that can enhance or decrease the innate immune response to LPS. CaOx promoted M1 macrophage development. These results suggest that monocytes have an important role promoting CaOx-induced inflammation.


Subject(s)
Calcium Oxalate/metabolism , Cell Differentiation , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Biomarkers , Cell Differentiation/immunology , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophage Activation/immunology , Macrophages/immunology , Monocytes/immunology , Nephrolithiasis/etiology , Nephrolithiasis/metabolism , Nephrolithiasis/pathology , Phagocytosis/immunology , THP-1 Cells
13.
Proc Natl Acad Sci U S A ; 114(5): 1117-1122, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096371

ABSTRACT

In recent years, it has been established that programmed cell death protein ligand 1 (PD-L1)-mediated inhibition of activated PD-1+ T lymphocytes plays a major role in tumor escape from immune system during cancer progression. Lately, the anti-PD-L1 and -PD-1 immune therapies have become an important tool for treatment of advanced human cancers, including bladder cancer. However, the underlying mechanisms of PD-L1 expression in cancer are not fully understood. We found that coculture of murine bone marrow cells with bladder tumor cells promoted strong expression of PD-L1 in bone marrow-derived myeloid cells. Tumor-induced expression of PD-L1 was limited to F4/80+ macrophages and Ly-6C+ myeloid-derived suppressor cells. These PD-L1-expressing cells were immunosuppressive and were capable of eliminating CD8 T cells in vitro. Tumor-infiltrating PD-L1+ cells isolated from tumor-bearing mice also exerted morphology of tumor-associated macrophages and expressed high levels of prostaglandin E2 (PGE2)-forming enzymes microsomal PGE2 synthase 1 (mPGES1) and COX2. Inhibition of PGE2 formation, using pharmacologic mPGES1 and COX2 inhibitors or genetic overexpression of PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), resulted in reduced PD-L1 expression. Together, our study demonstrates that the COX2/mPGES1/PGE2 pathway involved in the regulation of PD-L1 expression in tumor-infiltrating myeloid cells and, therefore, reprogramming of PGE2 metabolism in tumor microenvironment provides an opportunity to reduce immune suppression in tumor host.


Subject(s)
B7-H1 Antigen/biosynthesis , Bone Marrow Cells/metabolism , Cyclooxygenase 2/physiology , Dinoprostone/physiology , Macrophages/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Prostaglandin-E Synthases/physiology , Animals , B7-H1 Antigen/genetics , Cell Communication , Cell Line, Tumor , Coculture Techniques , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/genetics , Mice , Mice, Inbred C3H , Mice, Inbred NOD , Mice, SCID , Prostaglandin Antagonists/pharmacology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
14.
J Urol ; 195(4 Pt 1): 1143-51, 2016 04.
Article in English | MEDLINE | ID: mdl-26626217

ABSTRACT

PURPOSE: In murine and human hyperoxaluric conditions macrophages can be seen surrounding renal calcium oxalate crystal deposits. We hypothesized that macrophages have a role in degrading and destroying these deposits. We investigated the inflammatory response and phagocytic mechanisms when macrophages were exposed to human kidney stones and inorganic crystals. MATERIALS AND METHODS: Human monocytes were differentiated into resting, fully differentiated macrophages by treatment with recombinant human macrophage colony-stimulating factor (M-CSF) or GM-CSF (granulocyte M-CSF) for 6 days. After confirming phenotype by flow cytometry the macrophages were exposed for 20 hours to fragments of sterile human calcium oxalate stones or calcium oxalate crystals. Crystal uptake was determined, and supernatant cytokine and chemokine profiles were analyzed using antibody arrays. Quantitative reverse transcriptase-polymerase chain reaction was done to validate mRNA profile expression. RESULTS: Under direct vision fluorescence microscopy activated human macrophages were noted to surround stone fragments and synthesized crystals, and destroy them in a step-by-step process that involved clathrin mediated endocytosis and phagocytosis. An inflammatory cascade was released by macrophages, including the chemokines chemokine ligand (CCL)2, CCL3, interleukin (IL)-1 receptor antagonist (IL-1ra), complement component C5/C5a and IL-8. Response patterns to stone and crystal material depended on macrophage phenotype and activation status. CONCLUSIONS: In our in vitro study macrophages differentiated with M-CSF showed greater ability to phagocytize crystal deposits than those treated with GM-CSF. Following clathrin mediated endocytosis macrophages released a number of cytokines that are crucial for the inflammatory immune response. This suggests that tissue macrophages have an important role in preventing kidney stone disease by removing and digesting interstitial renal crystal deposits.


Subject(s)
Kidney Calculi/metabolism , Macrophages/metabolism , Phagocytosis/physiology , Calcium Oxalate/metabolism , Cell Culture Techniques , Chemokines/metabolism , Clathrin , Cytokines/metabolism , Flow Cytometry , Humans , Inflammation , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/physiology , Real-Time Polymerase Chain Reaction
15.
Clin Cancer Res ; 19(7): 1670-80, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23363815

ABSTRACT

PURPOSE: Chemokines are involved in cancer-related inflammation and malignant progression. In this study, we evaluated expression of CCR8 and its natural cognate ligand CCL1 in patients with urothelial carcinomas of bladder and renal cell carcinomas. EXPERIMENTAL DESIGN: We examined CCR8 expression in peripheral blood and tumor tissues from patients with bladder and renal carcinomas. CCR8-positive myeloid cells were isolated from cancer tissues with magnetic beads and tested in vitro for cytokine production and ability to modulate T-cell function. RESULTS: We show that monocytic and granulocytic myeloid cell subsets in peripheral blood of patients with cancer with urothelial and renal carcinomas display increased expression of chemokine receptor CCR8. Upregulated expression of CCR8 is also detected within human cancer tissues and primarily limited to tumor-associated macrophages. When isolated, CD11b(+)CCR8(+) cell subset produces the highest levels of proinflammatory and proangiogenic factors among intratumoral CD11b myeloid cells. Tumor-infiltrating CD11b(+)CCR8(+) cells selectively display activated Stat3 and are capable of inducing FoxP3 expression in autologous T lymphocytes. Primary human tumors produce substantial amounts of the natural CCR8 ligand CCL1. CONCLUSIONS: This study provides the first evidence that CCR8(+) myeloid cell subset is expanded in patients with cancer. Elevated secretion of CCL1 by tumors and increased presence of CCR8(+) myeloid cells in peripheral blood and cancer tissues indicate that CCL1/CCR8 axis is a component of cancer-related inflammation and may contribute to immune evasion. Obtained results also implicate that blockade of CCR8 signals may provide an attractive strategy for therapeutic intervention in human urothelial and renal cancers.


Subject(s)
Carcinoma/metabolism , Kidney Neoplasms/metabolism , Myeloid Cells/metabolism , Receptors, CCR8/metabolism , Urinary Bladder Neoplasms/metabolism , CD11b Antigen/metabolism , Carcinoma/pathology , Chemokine CCL1/metabolism , Humans , Inflammation/metabolism , Kidney Neoplasms/pathology , Leukocytes, Mononuclear , Urinary Bladder Neoplasms/pathology
16.
Oncoimmunology ; 1(2): 249-251, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22720260

ABSTRACT

Macrophage infiltration is a hallmark in the majority of solid tumors. Our studies demonstrated that macrophages that infiltrate human renal cells carcinoma (RCC) display markedly enhanced expression and activity of 15-lipoxygenase-2 (15-LOX2). Obtained data suggest that enhanced lipoxygenase activity in tumor-associated macrophages stimulates cancer inflammation and causes immune dysfunction.

17.
Int J Cancer ; 130(5): 1109-19, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21480223

ABSTRACT

Both cancer-related inflammation and tumor-induced immune suppression are associated with expansion of myeloid cell subsets including myeloid-derived suppressor cells. However, little known regarding characteristics of myeloid cells in patients with bladder cancer. In this study, we analyzed myeloid cells from peripheral blood (PBMC) and tumor tissue that were collected from patients with superficial noninvasive and invasive urothelial carcinomas. Our results demonstrate that PBMC from bladder cancer patients contain two major CD11b myeloid cell subsets: granulocyte-type CD15(high) CD33(low) cells and monocyte-type CD15(low) CD33(high) cells. The number of circulating granulocytic but not monocytic myeloid cells in cancer patients was markedly increased when compared to healthy individuals. Both myeloid cell subsets from cancer patients were highly activated and produced substantial amounts of proinflammatory chemokines/cytokines including CCL2, CCL3, CCL4, G-CSF, IL-8 and IL-6. Granulocytic myeloid cells were able to inhibit in vitro T cell proliferation through induction of CD4(+) Foxp3(+) T regulatory cells. Analysis of bladder cancer tissues revealed that tumors were infiltrated with monocyte-macrophage CD11b(+) HLA-DR(+) and granulocytic CD11b(+) CD15(+) HLA-DR(-) myeloid cells. Collectively, this study identifies myeloid cell subsets in patients with bladder cancer. We demonstrate that these highly activated inflammatory myeloid cells represent a source of multiple chemokines/cytokines and may contribute to inflammation and immune dysfunction in bladder cancer.


Subject(s)
Myeloid Cells/immunology , Urinary Bladder Neoplasms/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD11 Antigens/metabolism , Cytokines/metabolism , Granulocytes/immunology , Humans , Immune Tolerance , Lewis X Antigen/metabolism , Lymphocyte Activation , Monocytes/immunology , Sialic Acid Binding Ig-like Lectin 3
18.
Cancer Res ; 71(20): 6400-9, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21900394

ABSTRACT

Renal cell carcinoma (RCC), the most common human kidney cancer, is frequently infiltrated with tumor-associated macrophages (TAM) that can promote malignant progression. Here, we show that TAMs isolated from human RCC produce substantial amounts of the proinflammatory chemokine CCL2 and immunosuppressive cytokine IL-10, in addition to enhanced eicosanoid production via an activated 15-lipoxygenase-2 (15-LOX2) pathway. TAMs isolated from RCC tumors had a high 15-LOX2 expression and secreted substantial amounts of 15(S)-hydroxyeicosatetraenoic acid, its major bioactive lipid product. Inhibition of lipoxygenase activity significantly reduced production of CCL2 and IL-10 by RCC TAMs. In addition, TAMs isolated from RCC were capable of inducing in T lymphocytes, the pivotal T regulatory cell transcription factor forkhead box P3 (FOXP3), and the inhibitory cytotoxic T-lymphocyte antigen 4 (CTLA-4) coreceptor. However, this TAM-mediated induction of FOXP3 and CTLA-4 in T cells was independent of lipoxygenase and could not be reversed by inhibiting lipoxygenase activity. Collectively, our results show that TAMs, often present in RCCs, display enhanced 15-LOX2 activity that contributes to RCC-related inflammation, immunosuppression, and malignant progression. Furthermore, we show that TAMs mediate the development of immune tolerance through both 15-LOX2-dependent and 15-LOX2-independent pathways. We propose that manipulating LOX-dependent arachidonic acid metabolism in the tumor microenvironment could offer new strategies to block cancer-related inflammation and immune escape in patients with RCC.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Carcinoma, Renal Cell/enzymology , Immune Tolerance , Kidney Neoplasms/enzymology , Macrophages/enzymology , Aged , Arachidonate 15-Lipoxygenase/immunology , CTLA-4 Antigen/biosynthesis , CTLA-4 Antigen/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/surgery , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL2/immunology , Cyclooxygenase Inhibitors/pharmacology , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Interleukin-10/biosynthesis , Interleukin-10/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/surgery , Lipoxygenase Inhibitors/pharmacology , Macrophages/immunology , Male , Masoprocol/pharmacology , Middle Aged , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology
19.
Int Immunopharmacol ; 11(7): 848-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21315786

ABSTRACT

Bladder cancer is associated with enhanced inflammation and characterized by deregulated prostanoid metabolism. Here we examined prostaglandin E2 (PGE2) metabolism and myeloid cell subsets that infiltrate tumor tissue using two xenograft models of human bladder cancer. Human bladder tumor xenografts implanted into athymic nude mice become highly infiltrated with host CD11b myeloid cells of bone marrow origin. Fast growing SW780 bladder tumor xenografts were infiltrated with heterogeneous CD11b myeloid cell subsets including tumor-associated macrophages and myeloid-derived suppressor cells. In contrast, majority of myeloid cells in tumor tissue from slow growing bladder cancer Urothel 11 displayed more immature, homogenous phenotype and comprised mostly MHC II class-negative myeloid-derived suppressor cells. We demonstrate that human bladder tumors secrete substantial amounts of PGE2. Normal bone marrow myeloid cell progenitors cultured in the presence of a bladder tumor-conditioned medium, which is enriched for PGE2, failed to differentiate into mature APCs and acquired phenotype of the myeloid-derived suppressor cells or inflammatory macrophages with up-regulated chemokine receptor CXCR4. Collectively our data demonstrate that enhanced cancer-related inflammation and deregulated PGE2 metabolism in tumor microenvironment promote immunosuppressive pro-tumoral phenotype of myeloid cells in bladder cancer. These data also suggest that not only local tumor microenvironment but other factors such as stage of cancer disease and pace of tumor growth could markedly influence the phenotype, differentiation and immune function of myeloid cells in tumor tissue.


Subject(s)
Carcinoma/immunology , Dinoprostone/metabolism , Myeloid Cells/metabolism , Transplantation, Heterologous/pathology , Urinary Bladder Neoplasms/immunology , Animals , CD11b Antigen/biosynthesis , Carcinoma/pathology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Movement/immunology , Dinoprostone/immunology , Humans , Immunosuppression Therapy , Mice , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/pathology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Tumor Escape , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/pathology
20.
J Leukoc Biol ; 88(5): 839-48, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20587738

ABSTRACT

Recent studies suggest that tumor-infiltrated myeloid cells frequently up-regulate COX-2 expression and have enhanced PGE2 metabolism. This may affect the maturation and immune function of tumor-infiltrated antigen-presenting cells. In vitro studies demonstrate that tumor-derived factors can skew GM-CSF-driven differentiation of T(h)1-oriented myeloid APCs into M2-oriented Ly6C(+)F4/80(+) MDSCs or Ly6C(-)F4/80(+) arginase-expressing macrophages. These changes enable myeloid cells to produce substantial amounts of IL-10, VEGF, and MIP-2. The tumor-mediated inhibition of APC differentiation was associated with the up-regulated expression of PGE2-forming enzymes COX-2, mPGES1 in myeloid cells, and the simultaneous repression of PGE(2)-catabolizing enzyme 15-PGDH. The presence of tumor-derived factors also led to a reduced expression of PGT but promoted the up-regulation of MRP4, which works as a PGE2 efflux receptor. Addition of COX-2 inhibitor to the BM cell cultures could prevent the tumor-induced skewing of myeloid cell differentiation, partially restoring cell phenotype and down-regulating the arginase expression in the myeloid APCs. Our study suggests that tumors impair the intracellular PGE(2) catabolism in myeloid cells through simultaneous stimulation of PGE(2)-forming enzymes and inhibition of PGE2-degrading systems. This tumor-induced dichotomy drives the development of M2-oriented, arginase-expressing macrophages or the MDSC, which can be seen frequently among tumor-infiltrated myeloid cells.


Subject(s)
Colonic Neoplasms/pathology , Dinoprostone/metabolism , Macrophages/physiology , Myeloid Cells/immunology , Animals , Arginase/metabolism , Bone Marrow Cells/physiology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL