7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Fatty acid transporter 2 reprograms neutrophils in cancer

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Polymorphonuclear myeloid derived suppressor cells (PMN-MDSC) are pathologically activated neutrophils that are critically important for the regulation of immune responses in cancer. They contribute to the failure of cancer therapies and are associated with poor clinical outcomes. Despite the recent advances in understanding of the PMN-MDSC biology, the mechanisms responsible for pathological activation of neutrophils are not well defined, which limits selective targeting of these cells. Here, we report that mouse and human PMN-MDSC exclusively up-regulate fatty acid transporter protein 2 (FATP2). Over-expression of FATP2 in PMN-MDSC was controlled by GM-CSF, through the activation of STAT5 transcription factor. Deletion of FATP2 abrogated the suppressive activity of PMN-MDSC. The main mechanism of FATP2 mediated suppressive activity involved uptake of arachidonic acid (AA) and synthesis of prostaglandin E2 (PGE2). The selective pharmacological inhibition of FATP2 abrogated the activity of PMN-MDSC and substantially delayed tumor progression. In combination with check-point inhibitors it blocked tumor progression in mice. Thus, FATP2 mediates acquisition of immune suppressive activity by PMN-MDSC and represents a new target to selectively inhibit the functions of PMN-MDSC and improve the effect of cancer therapy.

          Related collections

          Most cited references19

          • Record: found
          • Abstract: found
          • Article: not found

          ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis.

          Dendritic cells (DCs) are required to initiate and sustain T cell-dependent anti-cancer immunity. However, tumors often evade immune control by crippling normal DC function. The endoplasmic reticulum (ER) stress response factor XBP1 promotes intrinsic tumor growth directly, but whether it also regulates the host anti-tumor immune response is not known. Here we show that constitutive activation of XBP1 in tumor-associated DCs (tDCs) drives ovarian cancer (OvCa) progression by blunting anti-tumor immunity. XBP1 activation, fueled by lipid peroxidation byproducts, induced a triglyceride biosynthetic program in tDCs leading to abnormal lipid accumulation and subsequent inhibition of tDC capacity to support anti-tumor T cells. Accordingly, DC-specific XBP1 deletion or selective nanoparticle-mediated XBP1 silencing in tDCs restored their immunostimulatory activity in situ and extended survival by evoking protective type 1 anti-tumor responses. Targeting the ER stress response should concomitantly inhibit tumor growth and enhance anti-cancer immunity, thus offering a unique approach to cancer immunotherapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Lipid accumulation and dendritic cell dysfunction in cancer

            Professional antigen presenting cells, dendritic cells (DC) are responsible for initiation and maintenance of immune responses. Here, we report that a substantial proportion of DCs in tumor-bearing mice and cancer patients have increased levels of triglycerides. Lipid accumulation in DCs was caused by increased uptake of extracellular lipids due to up-regulation of scavenger receptor A. DCs with high lipid content were not able to effectively stimulate allogeneic T cells or present tumor-associated antigens. DCs with high and normal lipid levels did not differ in expression of MHC and co-stimulatory molecules. However, lipid-laden DCs had reduced capacity to process antigens. Pharmacological normalization of lipid levels in DCs with an inhibitor of acetyl-CoA carboxylase restored the functional activity of DCs and substantially enhanced the effects of a cancer vaccine. These findings support the regulation of immune responses in cancer by manipulation of lipid levels in DCs.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma

              Myeloid suppressor cells (MSCs) producing high levels of arginase I block T cell function by depleting l-arginine in cancer, chronic infections, and trauma patients. In cancer, MSCs infiltrating tumors and in circulation are an important mechanism for tumor evasion and impair the therapeutic potential of cancer immunotherapies. However, the mechanisms that induce arginase I in MSCs in cancer are unknown. Using the 3LL mouse lung carcinoma, we aimed to characterize these mechanisms. Arginase I expression was independent of T cell–produced cytokines. Instead, tumor-derived soluble factors resistant to proteases induced and maintained arginase I expression in MSCs. 3LL tumor cells constitutively express cyclooxygenase (COX)-1 and COX-2 and produce high levels of PGE2. Genetic and pharmacological inhibition of COX-2, but not COX-1, blocked arginase I induction in vitro and in vivo. Signaling through the PGE2 receptor E-prostanoid 4 expressed in MSCs induced arginase I. Furthermore, blocking arginase I expression using COX-2 inhibitors elicited a lymphocyte-mediated antitumor response. These results demonstrate a new pathway of prostaglandin-induced immune dysfunction and provide a novel mechanism that can help explain the cancer prevention effects of COX-2 inhibitors. Furthermore, an addition of arginase I represents a clinical approach to enhance the therapeutic potential of cancer immunotherapies.
                Bookmark

                Author and article information

                Journal
                0410462
                6011
                Nature
                Nature
                Nature
                0028-0836
                1476-4687
                10 March 2019
                17 April 2019
                May 2019
                17 October 2019
                : 569
                : 7754
                : 73-78
                Affiliations
                [1– ]Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104;
                [2– ]Department of Environmental and Occupational Health, University of Pittsburgh, PA, 15219;
                [3– ]Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710;
                [4– ]Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104;
                [5– ]University of Pennsylvania School of Medicine, Philadelphia, PA, 19104,
                [6– ]Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, 19104
                [7– ]Department of Biochemistry, University of Nebraska, Lincoln, NE 68588
                [8– ]Helen F Graham Cancer Center at Christiana Care Health System, Wilmington, DE,
                [9– ]Departments of Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, USA, Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Russia
                Author notes

                Authors Contributions

                F.V. – participated in research design, performed most of the experiments, wrote manuscript, V.A.T. – performed lipidomics experiments, M.B. – prepared lentiviruses for experiments, A.D.L. – performed ChIP assay; L.D. – performed animal treatment experiments; A.K. – performed the analysis of RNAseq data; T.K.J. – performed some immunological experiments; Z.S. – performed metabolomics experiments, wrote manuscript, S.B. – performed seahorse experiments, F.W. – performed immunohistochemistry experiments; E.R. – generated COX2 KO cells; C.D. – generated lipofermata and wrote manuscript, M.E.M. – supervised metabolic experiments, reviewed manuscript, R.H.V. – supervised experiments with KPC mice, review manuscript, P.M.L. – supervised ChIP experiments, reviewed manuscript, C.M., B.N., N.H., G.M., M.G. – provided clinical samples; C.L. – generated mice with PMN targeted STAT5 deletion and performed in vivo experiments; Y.N. – generated mice with PMN targeted STAT5 deletion, reviewed manuscript, P.B. – produced lipofermata, reviewed manuscript, V. E. K. – obtained financial support for the study, designed experiments, supervised lipidomics analysis, wrote manuscript; D.I.G. – obtained financial support for the study, designed overall concept and specific experiments, supervised experiments, wrote manuscript.

                [10– ]Address for correspondence: The Wistar Institute, 3601 Spruce Str. Philadelphia, PA, 19104, dgabrilovich@ 123456wistar.org
                Article
                NIHMS1523620
                10.1038/s41586-019-1118-2
                6557120
                30996346
                4eb705e8-ab00-4afd-a72c-f1ecc0ffd595

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Uncategorized
                Uncategorized

                Comments

                Comment on this article