Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Front Immunol ; 12: 754083, 2021.
Article in English | MEDLINE | ID: mdl-34712241

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Myeloid-Derived Suppressor Cells/enzymology , Neoplasm Proteins/physiology , AMP-Activated Protein Kinases/physiology , Adoptive Transfer , Animals , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Kinase/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Female , Lymphocyte Depletion , Lymphoma/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Myeloid-Derived Suppressor Cells/physiology , Myeloid-Derived Suppressor Cells/transplantation , Myelopoiesis , Reactive Oxygen Species , Tumor Microenvironment
2.
Front Immunol ; 12: 690207, 2021.
Article in English | MEDLINE | ID: mdl-34149732

ABSTRACT

Histone deacetylase inhibitors (HDACIs) are antitumor drugs that are being developed for use in clinical settings. HDACIs enhance histone or nonhistone acetylation and promote gene transcription via epigenetic regulation. Importantly, these drugs have cytotoxic or cytostatic properties and can directly inhibit tumor cells. However, how HDACIs regulate immunocytes in the tumor microenvironment, such as myeloid-derived suppressor cells (MDSCs), has yet to be elucidated. In this review, we summarize the effects of different HDACIs on the immunosuppressive function and expansion of MDSCs based on the findings of relevant studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Neoplasms/drug therapy , Tumor Microenvironment , Acetylation , Animals , Antineoplastic Agents/adverse effects , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/adverse effects , Histones/metabolism , Humans , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/immunology
3.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074794

ABSTRACT

The DNA-sensing enzyme cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) regulates inflammation and immune defense against pathogens and malignant cells. Although cGAS has been shown to exert antitumor effects in several mouse models harboring transplanted tumor cell lines, its role in tumors arising from endogenous tissues remains unknown. Here, we show that deletion of cGAS in mice exacerbated chemical-induced colitis and colitis-associated colon cancer (CAC). Interestingly, mice lacking cGAS were more susceptible to CAC than those lacking stimulator of interferon genes (STING) or type I interferon receptor under the same conditions. cGAS but not STING is highly expressed in intestinal stem cells. cGAS deficiency led to intestinal stem cell loss and compromised intestinal barrier integrity upon dextran sodium sulfate-induced acute injury. Loss of cGAS exacerbated inflammation, led to activation of STAT3, and accelerated proliferation of intestinal epithelial cells during CAC development. Mice lacking cGAS also accumulated myeloid-derived suppressive cells within the tumor, displayed enhanced Th17 differentiation, but reduced interleukin (IL)-10 production. These results indicate that cGAS plays an important role in controlling CAC development by defending the integrity of the intestinal mucosa.


Subject(s)
Colonic Neoplasms/enzymology , Intestinal Mucosa/enzymology , Neoplasm Proteins/metabolism , Nucleotidyltransferases/metabolism , Animals , Colonic Neoplasms/genetics , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/enzymology , Neoplasm Proteins/genetics , Nucleotidyltransferases/genetics , Stem Cells/enzymology , Th17 Cells/enzymology
4.
Front Immunol ; 11: 574839, 2020.
Article in English | MEDLINE | ID: mdl-33250891

ABSTRACT

Background: Acetaminophen (APAP) overdose is one of the major etiologies of liver failure. Hepatocyte necrosis induced by toxic metabolites of APAP can activate proinflammatory responses, including elastase-expressing neutrophils, to exacerbate liver injury. Myeloid-derived suppressor cells (MDSCs) increased in inflammation can inhibit proinflammatory responses. Our aim is to investigate the role of MDSC in APAP-induced liver failure and the possible therapeutic application. Methods: BLAB/c mice were injected with a sublethal/lethal dose of APAP as the murine model of liver failure. MDSCs were defined as CD11b+Gr-1+ cells with the ability of T-cell suppression. Results: A sublethal challenge of APAP could increase the intrahepatic MDSC and protect mice against subsequent lethal challenge of APAP, lipopolysaccharide (LPS)/D-galatosamine or concanavalin A. This protection was lost if MDSCs were depleted and inducible nitric oxide synthase (iNOS) was the key molecule in this MDSC-mediated protection. Taking advantage of these observations, different bone marrow-derived MDSCs (BM-MDSCs) were generated. Among different cytokine-treated BM-MDSCs, tumor necrosis factor alpha/LPS-primed MDSCs (TNF-α/LPS MDSCs) had the strongest liver-protection ability after adoptive transfer. Further mechanistic explorations showed, iNOS-expressing TNF-α/LPS MDSCs induced the apoptosis of activated neutrophil and decreased the intrahepatic infiltration of elastase-expressing neutrophil. Moreover, we generated MDSCs from human peripheral blood mononuclear cells (PBMCs) with similar phenotype. Conclusion: We demonstrated the protective role of MDSCs and therapeutic effect of TNF-α/LPS MDSCs in APAP-induced liver failure. MDSC might protect against the APAP-induced liver failure by reducing the intrahepatic infiltration of activated neutrophil to limit inflammation. Therefore, a therapeutic role of MDSCs for APAP-induced liver failure was proposed.


Subject(s)
Adoptive Transfer , Chemical and Drug Induced Liver Injury/therapy , Liver Failure/therapy , Liver/enzymology , Myeloid-Derived Suppressor Cells/transplantation , Nitric Oxide Synthase Type II/metabolism , Acetaminophen , Animals , Apoptosis , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Humans , Leukocyte Elastase/metabolism , Liver/pathology , Liver Failure/chemically induced , Liver Failure/enzymology , Liver Failure/pathology , Male , Mice, Inbred BALB C , Mice, Knockout , Myeloid-Derived Suppressor Cells/enzymology , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Nitric Oxide Synthase Type II/genetics , Phenotype
5.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32530032

ABSTRACT

A homogeneous polysaccharide (GLP), with an average molecular weight of 4.44 × 104 Da, was isolated and purified from the fruiting bodies of Ganoderma lucidum. In this work, we examined the antitumor activities of GLP using a mouse Lewis lung cancer (LLC) model and explored possible molecular pathways involved in its immunomodulatory mechanism on tumor-host interaction. GLP administration (25 and 100 mg/kg) significantly inhibited tumor growth, as evidenced by the decreased tumor volume and tumor weight, as well as histological features of tumor tissues with concomitant down-regulation of proliferating cell nuclear antigen (PCNA) proliferative marker. Less myeloid-derived suppressor cells (MDSCs) were accumulated in both spleen and tumor tissues from GLP-treated mice. In contrast, the percentage of CD4+ and CD8+ T cells together with the production of Th1-type cytokines (IFN-γ and IL-12) was increased in the spleen of LLC-bearing mice following GLP administration. Furthermore, GLP administration reversed the attenuated expression of CARD9, p-Syk and p-p65, and increased indoleamine 2,3-dioxygenase (IDO) protein expression in MDSCs of LLC-bearing mice. Collectively, our data demonstrated the first time that GLP induced the differentiation of MDSCs and inhibited the accumulation of MDSCs via CARD9-NF-κB-IDO pathway, thus prevented lung cancer development.


Subject(s)
Antineoplastic Agents/pharmacology , CARD Signaling Adaptor Proteins/metabolism , Carcinoma, Lewis Lung/drug therapy , Cell Differentiation/drug effects , Fungal Polysaccharides/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Reishi , Animals , Antineoplastic Agents/isolation & purification , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Fruiting Bodies, Fungal , Fungal Polysaccharides/isolation & purification , Male , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/immunology , NF-kappa B/metabolism , Reishi/chemistry , Signal Transduction , Tumor Burden/drug effects , Tumor Microenvironment
6.
Exp Parasitol ; 215: 107917, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32446699

ABSTRACT

Cystic echinococcosis (CE) is a worldwide hazardous zoonotic parasitosis caused by Echinococcus granulosus. CE development involves complex immunological mechanisms, including participation of multiple immune cells and effector molecules. Myeloid-derived suppressor cells (MDSCs) are known to be involved in chronic and acute inflammatory conditions. In this study, we aimed to characterize the immune function of MDSCs in CE to improve the understanding, prevention and treatment of CE. Our results indicated that MDSCs overexpressing Ly6C and Ly6G inhibit the formation and activity of T helper 2 cells in a NO-dependent manner during E. granulosus infection.


Subject(s)
Echinococcosis/immunology , Echinococcus granulosus/immunology , Myeloid-Derived Suppressor Cells/immunology , Th2 Cells/immunology , Analysis of Variance , Animals , Antibodies, Monoclonal , Arginase/analysis , Bone Marrow/drug effects , Bone Marrow/immunology , Cytokines/analysis , Female , Flow Cytometry , Humans , Keratolytic Agents/pharmacology , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/enzymology , Nitric Oxide/analysis , Reactive Oxygen Species/analysis , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Tretinoin/pharmacology
7.
Clin Epigenetics ; 12(1): 13, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31941522

ABSTRACT

BACKGROUND: Increased numbers of myeloid-derived suppressor cells (MDSCs) are positively correlated with poor prognosis and reduced survivals of cancer patients. They play central roles in tumor immune evasion and tumor metastasis. However, limited data are available on phenotypic/transcriptomic characteristics of the different MDSCs subsets in cancer. These cells include immature (I-MDSCs), monocytic (M-MDSCs), and polymorphonuclear/granulocytic (PMN-MDSCs). METHODS: Phenotypic characterization of myeloid subsets from 27 colorectal cancer (CRC) patients was assessed by flow cytometric analyses. RNA-sequencing of sorted I-MDSCs, PMN-MDSCs, and antigen-presenting cells (APCs) was also performed. RESULTS: We found that the levels of I-MDSCs and PMN-MDSCs were increased in tumor tissues (TT), compared with normal tissues (NT) in colorectal cancer. Our functional annotation analyses showed that genes associated with histone deacetylase (HDAC) activation- and DNA methylation-mediated transcriptional silencing were upregulated, and histone acetyl transferase (HAT)-related genes were downregulated in tumor-infiltrating I-MDSCs. Moreover, pathways implicated in cell trafficking and immune suppression, including Wnt, interleukin-6 (IL-6), and mitogen-activated protein kinase (MAPK) signaling, were upregulated in I-MDSCs. Notably, PMN-MDSCs showed downregulation in genes related to DNA methylation and HDAC binding. Using an ex vivo model, we found that inhibition of HDAC activation or neutralization of IL-6 in CRC tumor tissues downregulates the expression of genes associated with immunosuppression and myeloid cell chemotaxis, confirming the importance of HDAC activation and IL-6 signaling pathway in MDSC function and chemotaxis. CONCLUSIONS: This study provides novel insights into the epigenetic regulations and other molecular pathways in different myeloid cell subsets within the CRC tumor microenvironment (TME), giving opportunities to potential targets for therapeutic benefits.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histone Code , Myeloid-Derived Suppressor Cells/metabolism , Adult , Aged , Aged, 80 and over , Cell Movement/genetics , Chemotaxis/genetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Gene Expression Profiling , Histone Deacetylases/metabolism , Humans , Immune Tolerance/genetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/physiology , Male , Middle Aged , Myeloid-Derived Suppressor Cells/enzymology
8.
Front Immunol ; 11: 579151, 2020.
Article in English | MEDLINE | ID: mdl-33537027

ABSTRACT

Graft-versus-host disease (GVHD) remains a major clinical drawback of allogeneic hematopoietic stem cell transplantation (HSCT). Here, we investigated how the stress responsive heme catabolizing enzyme heme oxygenase-1 (HO-1, encoded by HMOX1) regulates GVHD in response to allogeneic hematopoietic stem cell transplantation in mice and humans. We found that deletion of the Hmox1 allele, specifically in the myeloid compartment of mouse donor bone marrow, promotes the development of aggressive GVHD after allogeneic transplantation. The mechanism driving GVHD in mice transplanted with allogeneic bone marrow lacking HO-1 expression in the myeloid compartment involves enhanced T cell alloreactivity. The clinical relevance of these observations was validated in two independent cohorts of HSCT patients. Individuals transplanted with hematopoietic stem cells from donors carrying a long homozygous (GT)n repeat polymorphism (L/L) in the HMOX1 promoter, which is associated with lower HO-1 expression, were at higher risk of developing severe acute GVHD as compared to donors carrying a short (GT)n repeat (S/L or S/S) polymorphism associated with higher HO-1 expression. In this study, we showed the unique importance of donor-derived myeloid HO-1 in the prevention of lethal experimental GVHD and we corroborated this observation by demonstrating the association between human HMOX1 (GT)n microsatellite polymorphisms and the incidence of severe acute GVHD in two independent HSCT patient cohorts. Donor-derived myeloid HO-1 constitutes a potential therapeutic target for HSCT patients and large-scale prospective studies in HSCT patients are necessary to validate the HO-1 L/L genotype as an independent risk factor for developing severe acute GVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Myeloid-Derived Suppressor Cells/transplantation , Adult , Animals , Disease Models, Animal , Female , Genetic Predisposition to Disease , Graft vs Host Disease/enzymology , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Heme Oxygenase-1/genetics , Homozygote , Humans , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microsatellite Repeats , Middle Aged , Myeloid-Derived Suppressor Cells/enzymology , Phenotype , Polymorphism, Genetic , Retrospective Studies , Risk Assessment , Risk Factors , Severity of Illness Index
9.
J Cell Physiol ; 234(2): 1618-1629, 2019 02.
Article in English | MEDLINE | ID: mdl-30132862

ABSTRACT

Accumulating evidence has shown that mammalian target of rapamycin (mTOR) pathway and myeloid-derived suppressor cells (MDSCs) are involved in pathogenesis of inflammatory bowel diseases (IBDs). INK128 is a novel mTOR kinase inhibitor in clinical development. However, the exact roles of MDSCs and INK128 in IBD are unclear. Here, we showed that the INK128 treatment enhanced the resistance of mice to dextran sodium sulfate (DSS)-induced colitis and inhibited the differentiation of MDSCs into macrophages. Moreover, interferon (IFN)-α level was elevated in INK128-treated colitis mice. When stimulated with IFN-α in vitro, MDSCs showed a superior immunosuppression activity. Of note, the regulatory T cells (Tregs) increased but Th1 cells decreased in INK128-treated colitis mice. These results indicate that mTOR inhibitor INK128 attenuates DSS-induced colitis via Treg expansion promoted by MDSCs. Our work provides a new evidence that INK128 is potential to be a therapeutic drug on DSS-induced colitis via regulating MDSCs as well as maintaining Treg expansion.


Subject(s)
Benzoxazoles/pharmacology , Cell Proliferation/drug effects , Colitis/prevention & control , Colon/drug effects , Lymphocyte Activation/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Communication/drug effects , Colitis/chemically induced , Colitis/enzymology , Colitis/immunology , Colon/enzymology , Colon/immunology , Dextran Sulfate , Disease Models, Animal , Female , Macrophages/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/immunology , RAW 264.7 Cells , Signal Transduction , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism
10.
FASEB J ; 32(5): 2339-2353, 2018 05.
Article in English | MEDLINE | ID: mdl-29259036

ABSTRACT

Bioactive sphingolipids are modulators of immune processes and their metabolism is often dysregulated in ulcerative colitis, a major category of inflammatory bowel disease (IBD). While multiple axes of sphingolipid metabolism have been investigated to delineate mechanisms regulating ulcerative colitis, the role of acid ceramidase (AC) in intestinal inflammation is yet to be characterized. Here we demonstrate that AC expression is elevated selectively in the inflammatory infiltrate in human and murine colitis. To probe for mechanistic insight into how AC up-regulation can impact intestinal inflammation, we investigated the selective loss of AC expression in the myeloid population. Using a model of intestinal epithelial injury, we demonstrate that myeloid AC conditional knockout mice exhibit impairment of neutrophil recruitment to the colon mucosa as a result of defective cytokine and chemokine production. Furthermore, the loss of myeloid AC protects from tumor incidence in colitis-associated cancer (CAC) and inhibits the expansion of neutrophils and granulocytic myeloid-derived suppressor cells in the tumor microenvironment. Collectively, our results demonstrate a tissue-specific role for AC in regulating neutrophilic inflammation and cytokine production. We demonstrate novel mechanisms of how granulocytes are recruited to the colon that may have therapeutic potential in intestinal inflammation, IBD, and CAC.-Espaillat, M. P., Snider, A. J., Qiu, Z., Channer, B., Coant, N., Schuchman, E. H., Kew, R. R., Sheridan, B. S., Hannun, Y. A., Obeid, L. M. Loss of acid ceramidase in myeloid cells suppresses intestinal neutrophil recruitment.


Subject(s)
Acid Ceramidase/biosynthesis , Colitis, Ulcerative/enzymology , Colon/enzymology , Gene Expression Regulation, Enzymologic , Intestinal Mucosa/enzymology , Neutrophils/enzymology , Up-Regulation , Acid Ceramidase/genetics , Animals , Chemokines/biosynthesis , Chemokines/genetics , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neutrophils/pathology , Tumor Microenvironment/genetics
11.
Biol Blood Marrow Transplant ; 24(1): 32-42, 2018 01.
Article in English | MEDLINE | ID: mdl-28844945

ABSTRACT

The recovery of myeloid-derived suppressor cells (MDSCs) and its relevance in clinical acute graft-versus-host disease (GVHD) and post-hematopoietic stem cell transplantation (HSCT) infections remain to be fully characterized. We examined the expansion of circulating monocytic (M-) MDSCs and granulocytic (G-) MDSCs at the time of engraftment in 130 patients undergoing allogeneic HSCT (allo-HSCT). Compared with the G-MDSC group, the high M-MDSC group had a higher infection rate within 100 days, along with worse 1-year cumulative incidence of treatment-related mortality (TRM) and 2-year probability of event-free survival (EFS). The frequency of M-MDSCs was associated with preceding severe mucositis. Transcriptome profiling analysis of 2 isolated MDSC subtype showed significantly greater matrix metalloproteinase-9 (MMP-9) expression in M-MDSCs than in G-MDSCs. M-MDSCs produced abundantly more MMP-9. Importantly, compared with G-MDSCs, M-MDSCs isolated from patients post-HSCT had a greater capacity to suppress T cell responses, and MMP-9 blockade more forcefully inhibited their immunosuppressive effect. MMP-9 levels also were associated with the occurrence of infections and with transplantation outcomes. Based on these findings, we identify M-MDSCs as a major contributor to infections early after allo-HSCT and worse clinical outcomes via MMP-9.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/enzymology , Transplantation, Homologous/adverse effects , Treatment Outcome , Adult , Female , Gene Expression Profiling , Graft vs Host Disease/etiology , Granulocytes/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infections/etiology , Male , Middle Aged , Monocytes/enzymology , Myeloid-Derived Suppressor Cells/pathology
12.
Nat Commun ; 8(1): 517, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894087

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) possess immunosuppressive activities, which allow cancers to escape immune surveillance and become non-responsive to immune checkpoints blockade. Here we report hypoxia as a cause of MDSC accumulation. Using hepatocellular carcinoma (HCC) as a cancer model, we show that hypoxia, through stabilization of hypoxia-inducible factor-1 (HIF-1), induces ectoenzyme, ectonucleoside triphosphate diphosphohydrolase 2 (ENTPD2/CD39L1), in cancer cells, causing its overexpression in HCC clinical specimens. Overexpression of ENTPD2 is found as a poor prognostic indicator for HCC. Mechanistically, we demonstrate that ENTPD2 converts extracellular ATP to 5'-AMP, which prevents the differentiation of MDSCs and therefore promotes the maintenance of MDSCs. We further find that ENTPD2 inhibition is able to mitigate cancer growth and enhance the efficiency and efficacy of immune checkpoint inhibitors. Our data suggest that ENTPD2 may be a good prognostic marker and therapeutic target for cancer patients, especially those receiving immune therapy.Myeloid-derived suppressor cells (MDSCs) promote tumor immune escape. Here, the authors show that in hepatocellular carcinoma, hypoxia induces the expression of ENTPD2 on cancer cells leading to elevated extracellular 5'-AMP, which in turn promote the maintenance of MDSCs by preventing their differentiation.


Subject(s)
Adenosine Triphosphatases/metabolism , Carcinoma, Hepatocellular/enzymology , Hypoxia-Inducible Factor 1/metabolism , Liver Neoplasms/enzymology , Myeloid-Derived Suppressor Cells/enzymology , Adenosine Triphosphatases/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/physiopathology , Cell Differentiation , Cell Proliferation , Humans , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism
13.
Oncotarget ; 7(51): 83907-83925, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27880732

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immune suppressive cells that are hallmarks of human cancer. MDSCs inhibit cytotoxic T lymphocytes (CTLs) and NK cell functions to promote tumor immune escape and progression, and therefore are considered key targets in cancer immunotherapy. Recent studies determined a key role of the apoptosis pathways in tumor-induced MDSC homeostasis and it is known that ceramide plays a key role in regulation of mammalian cell apoptosis. In this study, we aimed to determine the efficacy and underlying molecular mechanism of ceramide in suppression of MDSCs. Treatment of tumor-bearing mice with LCL521, a lysosomotropic inhibitor of acid ceramidase, significantly decreased MDSC accumulation in vivo. Using a MDSC-like myeloid cell model, we determined that LCL521 targets lysosomes and increases total cellular C16 ceramide level. Although MDSC-like cells have functional apoptosis pathways, LCL521-induced MDSC death occurs in an apoptosis- and necroptosis-independent mechanism. LCL521 treatment resulted in an increase in the number of autophagic vesicles, heterolysosomes and swollen ERs. Finally, concomitant inhibition of cathepsin B and cathepsin D was required to significantly decrease LCL521-induced cell death. Our observations indicate that LCL521 targets lysosomes to activate cathepsin B and cathepsin D, resulting in interrupted autophagy and ER stress that culminates in MDSC death. Therefore, a ceramidase inhibitor is potentially an effective adjunct therapeutic agent for suppression of MDSCs to enhance the efficacy of CTL-based cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cathepsin B/metabolism , Cathepsin D/metabolism , Ceramides/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Lysosomes/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Sarcoma/drug therapy , Signal Transduction/drug effects , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Lysosomes/enzymology , Lysosomes/pathology , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/pathology , Sarcoma/enzymology , Sarcoma/immunology , Sarcoma/pathology , Time Factors
14.
Sci Rep ; 6: 31397, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27507680

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are highly prevalent inflammatory cells that play a key role in tumor development and are considered therapeutic targets. MDSCs promote tumor growth by blocking T-cell-mediated anti-tumoral immune response through depletion of arginine that is essential for T-cell proliferation. To deplete arginine, MDSCs express high levels of arginase, which catalyzes the breakdown of arginine into urea and ornithine. Here, we developed a new hyperpolarized (13)C probe, [6-(13)C]-arginine, to image arginase activity. We show that [6-(13)C]-arginine can be hyperpolarized, and hyperpolarized [(13)C]-urea production from [6-(13)C]-arginine is linearly correlated with arginase concentration in vitro. Furthermore we show that we can detect a statistically significant increase in hyperpolarized [(13)C]-urea production in MDSCs when compared to control bone marrow cells. This increase was associated with an increase in intracellular arginase concentration detected using a spectrophotometric assay. Hyperpolarized [6-(13)C]-arginine could therefore serve to image tumoral MDSC function and more broadly M2-like macrophages.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Myeloid-Derived Suppressor Cells/cytology , Animals , Arginine/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , Cells, Cultured , Mice , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/metabolism , Urea/chemistry
15.
Cancer Res ; 76(8): 2125-36, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26880800

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of immature myeloid cells that expand in tumor-bearing hosts in response to soluble factors produced by tumor and stromal cells. MDSC expansion has been linked to loss of immune effector cell function and reduced efficacy of immune-based cancer therapies, highlighting the MDSC population as an attractive therapeutic target. Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK) and IL2-inducible T-cell kinase (ITK), is in clinical use for the treatment of B-cell malignancies. Here, we report that BTK is expressed by murine and human MDSCs, and that ibrutinib is able to inhibit BTK phosphorylation in these cells. Treatment of MDSCs with ibrutinib significantly impaired nitric oxide production and cell migration. In addition, ibrutinib inhibited in vitro generation of human MDSCs and reduced mRNA expression of indolamine 2,3-dioxygenase, an immunosuppressive factor. Treatment of mice bearing EMT6 mammary tumors with ibrutinib resulted in reduced frequency of MDSCs in both the spleen and tumor. Ibrutinib treatment also resulted in a significant reduction of MDSCs in wild-type mice bearing B16F10 melanoma tumors, but not in X-linked immunodeficiency mice (XID) harboring a BTK mutation, suggesting that BTK inhibition plays an important role in the observed reduction of MDSCs in vivo Finally, ibrutinib significantly enhanced the efficacy of anti-PD-L1 (CD274) therapy in a murine breast cancer model. Together, these results demonstrate that ibrutinib modulates MDSC function and generation, revealing a potential strategy for enhancing immune-based therapies in solid malignancies. Cancer Res; 76(8); 2125-36. ©2016 AACR.


Subject(s)
Myeloid-Derived Suppressor Cells/enzymology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis , Cell Line, Tumor , Cytokines/biosynthesis , Gene Expression , Humans , Immunotherapy , Mice , Piperidines
16.
Oncotarget ; 7(8): 8866-78, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26758420

ABSTRACT

Hepatic stellate cells (HSCs) are critical mediators of immunosuppression and the pathogenesis of hepatocellular carcinoma (HCC). Our previous work indicates that HSCs promote HCC progression by enhancing immunosuppressive cell populations including myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). MDSCs are induced by inflammatory cytokines (e.g., prostaglandins) and are important in immune suppression. However, how HSCs mediate expansion of MDSCs is uncertain. Thus, we studied activated HSCs that could induce MDSCs from bone marrow cells and noted that HSC-induced MDSCs up-regulated immunosuppressive activity via iNOS, Arg-1, and IL-4Rα. After treating cells with a COX-2 inhibitor or an EP4 antagonist, we established that HSC-induced MDSC accumulation was mediated by the COX2-PGE2-EP4 signaling. Furthermore, in vivo animal studies confirmed that inhibition of HSC-derived PGE2 could inhibit HSC-induced MDSC accumulation and HCC growth. Thus, our data show that HSCs are required for MDSC accumulation mediated by the COX2-PGE2-EP4 pathway, and these data are the first to link HSC and MDSC subsets in HCC immune microenvironment and provide a rationale for targeting PGE2 signaling for HCC therapy.


Subject(s)
Bone Marrow/pathology , Carcinoma, Hepatocellular/pathology , Cyclooxygenase 2/metabolism , Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Animals , Bone Marrow/drug effects , Bone Marrow/enzymology , Carcinoma, Hepatocellular/enzymology , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Flow Cytometry , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/enzymology , Liver Neoplasms/enzymology , Male , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/enzymology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL