Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Immunity ; 41(6): 947-59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500368

ABSTRACT

Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Granulocytes/physiology , Monocytes/physiology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells/physiology , Neoplasms, Experimental/immunology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Caspase 8/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/genetics , Coculture Techniques , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Signal Transduction/genetics
2.
Blood ; 126(13): 1621-8, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26265697

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a naturally occurring immune regulatory population associated with inhibition of ongoing inflammatory responses. In vitro generation of MDSCs from bone marrow has been shown to enhance survival in an acute model of lethal graft-versus-host disease (GVHD). However, donor MDSC infusion only partially ameliorates GVHD lethality. In order to improve the potential therapeutic benefit and ultimately survival outcomes, we set out to investigate the fate of MDSCs after transfer in the setting of acute GVHD (aGVHD). MDSCs transferred to lethally irradiated recipients of allogeneic donor hematopoietic grafts are exposed to an intense inflammatory environment associated with aGVHD, which we now show directly undermines their suppressive capacity. Under a conditioning regimen and GVHD inflammatory settings, MDSCs rapidly lose suppressor function and their potential to inhibit GVHD lethality, which is associated with their induced conversion toward a mature inflammasome-activated state. We find even brief in vitro exposure to inflammasome-activating mediators negates the suppressive potential of cultured murine and human-derived MDSCs. Consistent with a role for the inflammasome, donor MDSCs deficient in the adaptor ASC (apoptosis-associated speck-like protein containing a CARD), which assembles inflammasome complexes, conferred improved survival of mice developing GVHD compared with wild-type donor MDSCs. These data suggest the use of MDSCs as a therapeutic approach for preventing GVHD and other systemic inflammatory conditions will be more effective when combined with approaches limiting in vivo MDSC inflammasome activation, empowering MDSCs to maintain their suppressive potential.


Subject(s)
Adoptive Transfer , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Inflammasomes/immunology , Myeloid Cells/immunology , Myeloid Cells/transplantation , Animals , Bone Marrow Cells/cytology , CD11 Antigens/immunology , Cell Differentiation , Cells, Cultured , Graft vs Host Disease/pathology , Humans , Interleukin-1beta/immunology , Mice , Myeloid Cells/cytology , Myeloid Cells/pathology
3.
Eur J Immunol ; 41(3): 749-59, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21287554

ABSTRACT

Current paradigms suggest that, despite the heterogeneity of myeloid-derived suppressor cells (MDSC), all Gr-1(+) CD11b(+) cells can exert suppressive function when exposed to inflammatory stimuli. In vitro evaluation shows that MDSC from multiple tissue sites have suppressive activity, and in vivo inhibition of MDSC enhances T-cell function; however, the relative capacity of MDSC present at localized inflammatory sites or in peripheral tissues to suppress T-cell responses in vivo has not been directly evaluated. In the current study, we observed that during a tissue-specific inflammatory response, MDSC inhibition of CD8(+) T-cell proliferation and IFN-γ production was restricted to the inflammatory site. Using a prostate-specific inflammatory model and a heterotopic prostate tumor model, we showed that MDSC from inflammatory sites or from tumor tissue possess immediate capacity to inhibit T-cell function, whereas those isolated from peripheral tissues (spleens and liver) were not suppressive without activation of iNOS by exposure to IFN-γ. These data suggest that MDSC are important regulators of immune responses in the prostate during acute inflammation and the chronic inflammatory setting of tumor growth, and that regulation of T-cell function by MDSC during a localized inflammatory response is restricted in vivo to the site of an ongoing immune response.


Subject(s)
Immune Tolerance , Inflammation/immunology , Myeloid Cells/immunology , Adoptive Transfer , Animals , Arginase/genetics , CD11b Antigen/metabolism , Cell Proliferation , Disease Models, Animal , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Transgenic , Myeloid Cells/pathology , Nitric Oxide Synthase Type II/genetics , Ovalbumin/genetics , Ovalbumin/immunology , Phenotype , Prostatitis/genetics , Prostatitis/immunology , Prostatitis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chemokine/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
4.
Am J Pathol ; 179(1): 502-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703427

ABSTRACT

Prostate-specific deletion of Pten in mice has been reported to recapitulate histological progression of human prostate cancer. To improve on this model, we introduced the conditional ROSA26 luciferase reporter allele to monitor prostate cancer progression via bioluminescence imaging and extensively backcrossed mice onto the albino C57BL/6 genetic background to address variability in tumor kinetics and to enhance imaging sensitivity. Bioluminescence signal increased rapidly in Pten(p-/-) mice from 3 to 11 weeks, but was much slower from 11 to 52 weeks. Changes in bioluminescence signal were correlated with epithelial proliferation. Magnetic resonance imaging revealed progressive increases in prostate volume, which were attributed to excessive fluid retention in the anterior prostate and to expansion of the stroma. Development of invasive prostate cancer in 52-week-old Pten(p-/-) mice was rare, indicating that disease progression was slowed relative to that in previous reports. Tumors in these mice exhibited a spontaneous inflammatory phenotype and were rapidly infiltrated by myeloid-derived suppressor cells. Although Pten(p-/-) tumors responded to androgen withdrawal, they failed to exhibit relapsed growth for up to 1 year. Taken together, these data identify a mild prostate cancer phenotype in C57BL/6 prostate-specific Pten-deficient mice, reflecting effects of the C57BL/6 genetic background on cancer progression. This model provides a platform for noninvasive assessment of how genetic and environmental risk factors may affect disease progression.


Subject(s)
Disease Models, Animal , Inflammation/pathology , PTEN Phosphohydrolase/physiology , Prostate/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Animals , Cell Proliferation , Disease Progression , Fluorescent Antibody Technique , Humans , Integrases , Luciferases/metabolism , Lymphatic Metastasis , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Prostate/metabolism , Prostatic Neoplasms/genetics , Time Factors
5.
Immunohorizons ; 6(12): 790-806, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36480485

ABSTRACT

Owing to ease of access and high yield, most murine myeloid-derived suppressor cell (MDSC) knowledge comes from the study of spleen-derived MDSCs rather than those isolated from the tumor. Although several studies have identified subtle differences in suppressive function between these MDSCs, a recent report demonstrated that the whole peripheral myeloid compartment poorly reflects myeloid populations found at the tumor. We confirm and extend these observations by presenting data that indicate extensive differences exist between peripheral and tumor MDSCs, suggesting that it may be inappropriate to use spleen MDSCs as surrogates for studying tumor MDSCs. Using cytospins, we observed that tumor MDSCs have undergone a morphologic shift from immature myeloid cell forms commonly seen in bone marrow (BM) and spleen MDSCs and acquired mature myeloid cell characteristics. Spleen and BM monocyte-like MDSCs (M-MDSCs) readily responded to differentiation signals for multiple myeloid cell types whereas tumor M-MDSCs had remarkably reduced cellular plasticity. At the time of isolation, M-MDSCs from BM or spleen have little to no T cell suppressive activity whereas those from the tumor possess immediate and efficient T cell suppressive function. Finally, microarray analysis revealed that the transcriptomes of tumor and spleen M-MDSCs possessed >4500 differentially expressed transcripts. We conclude that tumor M-MDSCs are more differentiated and mature, and that they are morphologically, genetically, and functionally distinct from spleen and BM M-MDSCs. These observations have important implications for the design of anti-MDSC therapies and suggest that preclinical studies using nontumor MDSCs could lead to results not applicable to tumor MDSCs.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Animals , Mice , Monocytes , Cell Differentiation
6.
Prostate ; 71(11): 1139-50, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21656824

ABSTRACT

BACKGROUND: Prostatitis is a poorly understood disease and increasing evidence suggests inflammation is involved in other prostatic diseases including prostate cancer. METHODS: The ability of pre-activated CD8 T cells to induce prostatitis was examined by adoptive transfer of prostate antigen specific CD8 T cells into POET-3 mice or POET-3/Luc/Pten(-/+) mice. Characterization of the inflammatory response was determined by examining leukocyte infiltration by histological analysis, flow cytometry and by evaluating cytokine and chemokine levels in prostate tissue. The impact of inflammation on the prostate was evaluated by monitoring epithelial cell proliferation over time. RESULTS: Initiation of inflammation by ovalbumin specific CD8⁺ T cells (OT-I cells) resulted in development of acute prostatitis in the anterior, dorsolateral and ventral prostate of POET-3 and POET-3/Luc/Pten(-/+) mice. Acute prostatitis was characterized by recruitment of adoptively transferred OT-I cells and importantly, autologous CD4⁺ and CD8⁺ T cells, myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg). In concert with leukocyte infiltration elevated levels of pro-inflammatory cytokines and chemokines were observed. Inflammation also resulted in marked epithelial cell proliferation that was sustained up to 80 days post adoptive transfer of OT-I cells. CONCLUSIONS: The POET-3 model represents a novel mouse model to study both acute and chronic prostate inflammation in an antigen-specific system. Further, the POET-3 mouse model can be crossed with other genetic models of disease such as the C57/Luc/Pten(-/-) model of prostate cancer, allowing the impact of prostatitis on other prostatic diseases to be evaluated.


Subject(s)
Cell Proliferation , Disease Models, Animal , Epitopes/biosynthesis , Prostate/immunology , Prostate/pathology , Prostatitis/immunology , Prostatitis/pathology , Acute Disease , Animals , Chronic Disease , Epitopes/immunology , Inflammation/diagnosis , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
7.
Oncotarget ; 10(68): 7220-7237, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31921384

ABSTRACT

The synthetic oligonucleotide SD-101 is a potent and specific agonist for toll-like receptor 9. Intratumoral injection of SD-101 induces significant anti-tumor immunity in preclinical and clinical studies, especially when combined with PD-1 blockade. To build upon this strategy, we studied the enhancement of SD-101 activities by combination with low-dose cyclophosphamide, a well-characterized agent with potentially complementary activities. In multiple mouse tumor models, we demonstrate substantial anti-tumor activity of the combination, compared to each single agent. Combination therapy generated CD8+ T cell dependent immunity leading to rejection of both non-injected and injected tumors and long-term survival, even in very large tumors. Mechanistic studies encompassing global gene expression changes and characterization of immune cell infiltrates show the rapid, sequential induction of innate and adaptive responses and identify discrete contributions of SD-101 and cyclophosphamide. Importantly, these changes were prominent in tumors not injected directly with SD-101. Combination treatment resulted in creation of a permissive environment for a systemic anti-tumor immune response, including a reduction of intratumoral regulatory T cells (Tregs) and an increase in "M1" versus "M2" tumor-associated macrophage (TAM) phenotypes. Additionally, we observed increased immunogenic cell death as well as antigen processing in response to combination treatment.

8.
Cell Rep ; 12(11): 1902-14, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26365184

ABSTRACT

Cancer can involve non-resolving, persistent inflammation where varying numbers of tumor-associated macrophages (TAMs) infiltrate and adopt different activation states between anti-tumor M1 and pro-tumor M2 phenotypes. Here, we resolve a cascade causing differential macrophage phenotypes in the tumor microenvironment. Reduction in TNF mRNA production or loss of type I TNF receptor signaling resulted in a striking pattern of enhanced M2 mRNA expression. M2 gene expression was driven in part by IL-13 from eosinophils co-recruited with inflammatory monocytes, a pathway that was suppressed by TNF. Our data define regulatory nodes within the tumor microenvironment that balance M1 and M2 populations. Our results show macrophage polarization in cancer is dynamic and dependent on the balance between TNF and IL-13, thus providing a strategy for manipulating TAMs.


Subject(s)
Macrophages/metabolism , Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL