15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      ALKBH5 regulates anti–PD-1 therapy response by modulating lactate and suppressive immune cell accumulation in tumor microenvironment

      research-article

      Read this article at

      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.

          Significance

          N 6 -methylation of adenosine (m 6A) RNA modification plays important roles in development and tumorigenesis. The functions and mechanisms of m 6A demethylases during cancer immunotherapy is still unclear. Here we employed melanoma and colon syngeneic mouse models to study the roles of m 6A demethylases ALKBH5 and FTO during anti–PD-1 antibody and GVAX vaccination therapy. We found that ALKBH5 knockout in tumor cells enhances efficacy of immunotherapy and prolonged mouse survival. ALKBH5 modulates target gene expression and gene splicing, leading to changes of metabolite contents, such as lactate in tumor microenvironment, which regulates suppressive lymphocytes Treg and myeloid-derived suppressor cell accumulations. Importantly, by using ALKBH5-specific inhibitor, we observed the similar phenotype, indicating future translational application of our findings.

          Abstract

          Although immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment, many patients do not respond or develop resistance to ICB. N 6 -methylation of adenosine (m 6A) in RNA regulates many pathophysiological processes. Here, we show that deletion of the m 6A demethylase Alkbh5 sensitized tumors to cancer immunotherapy. Alkbh5 has effects on m 6A density and splicing events in tumors during ICB. Alkbh5 modulates Mct4/Slc16a3 expression and lactate content of the tumor microenvironment and the composition of tumor-infiltrating Treg and myeloid-derived suppressor cells. Importantly, a small-molecule Alkbh5 inhibitor enhanced the efficacy of cancer immunotherapy. Notably, the ALKBH5 gene mutation and expression status of melanoma patients correlate with their response to immunotherapy. Our results suggest that m 6A demethylases in tumor cells contribute to the efficacy of immunotherapy and identify ALKBH5 as a potential therapeutic target to enhance immunotherapy outcome in melanoma, colorectal, and potentially other cancers.

          Related collections

          Most cited references26

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

          Rethinking m6A Readers, Writers, and Erasers.

          In recent years, m6A has emerged as an abundant and dynamically regulated modification throughout the transcriptome. Recent technological advances have enabled the transcriptome-wide identification of m6A residues, which in turn has provided important insights into the biology and regulation of this pervasive regulatory mark. Also central to our current understanding of m6A are the discovery and characterization of m6A readers, writers, and erasers. Over the last few years, studies into the function of these proteins have led to important discoveries about the regulation and function of m6A. However, during this time our understanding of these proteins has also evolved considerably, sometimes leading to the reversal of early concepts regarding the reading, writing and erasing of m6A. In this review, we summarize recent advances in m6A research, and we highlight how these new findings have reshaped our understanding of how m6A is regulated in the transcriptome.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            m 6 A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade

            Melanoma is one of the most deadly and therapy-resistant cancers. Here we show that N6-methyladenosine (m6A) mRNA demethylation by fat mass and obesity-associated protein (FTO) increases melanoma growth and decreases response to anti-PD-1 blockade immunotherapy. FTO level is increased in human melanoma and enhances melanoma tumorigenesis in mice. FTO is induced by metabolic starvation stress through the autophagy and NF-κB pathway. Knockdown of FTO increases m6A methylation in the critical protumorigenic melanoma cell-intrinsic genes including PD-1 (PDCD1), CXCR4, and SOX10, leading to increased RNA decay through the m6A reader YTHDF2. Knockdown of FTO sensitizes melanoma cells to interferon gamma (IFNγ) and sensitizes melanoma to anti-PD-1 treatment in mice, depending on adaptive immunity. Our findings demonstrate a crucial role of FTO as an m6A demethylase in promoting melanoma tumorigenesis and anti-PD-1 resistance, and suggest that the combination of FTO inhibition with anti-PD-1 blockade may reduce the resistance to immunotherapy in melanoma.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              ALKBH5-dependent m6A demethylation controls splicing and stability of long 3'-UTR mRNAs in male germ cells.

              N6-methyladenosine (m6A) represents one of the most common RNA modifications in eukaryotes. Specific m6A writer, eraser, and reader proteins have been identified. As an m6A eraser, ALKBH5 specifically removes m6A from target mRNAs and inactivation ofAlkbh5leads to male infertility in mice. However, the underlying molecular mechanism remains unknown. Here, we report that ALKBH5-mediated m6A erasure in the nuclei of spermatocytes and round spermatids is essential for correct splicing and the production of longer 3'-UTR mRNAs, and failure to do so leads to aberrant splicing and production of shorter transcripts with elevated levels of m6A that are rapidly degraded. Our study identified reversible m6A modification as a critical mechanism of posttranscriptional control of mRNA fate in late meiotic and haploid spermatogenic cells.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                18 August 2020
                3 August 2020
                3 August 2020
                : 117
                : 33
                : 20159-20170
                Affiliations
                [1] aDivision of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego , La Jolla, CA 92093;
                [2] bBioinformatics Program, University of California San Diego , La Jolla, CA 92093;
                [3] cEnvironmental Toxicology Graduate Program, University of California, Riverside , CA 92521;
                [4] dDepartment of Chemistry, University of California, Riverside , CA 92521;
                [5] eSan Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California San Diego , La Jolla, CA 92093
                Author notes
                1To whom correspondence may be addressed. Email: trana@ 123456ucsd.edu .

                Edited by Stephen P. Goff, Columbia University Medical Center, New York, NY, and approved July 7, 2020 (received for review October 31, 2019)

                Author contributions: T.M.R. conceived and planned the project; N.L. and T.M.R. designed research; N.L., L.W., S.H., R.T., and G.M.G. performed research; S.P.P. contributed new reagents/analytic tools; N.L., Y.K., H.H., K.A., Y.W., S.P.P., and T.M.R. analyzed data; and N.L. and T.M.R. wrote the paper.

                Author information
                https://orcid.org/0000-0002-2852-8099
                Article
                201918986
                10.1073/pnas.1918986117
                7443867
                32747553
                e111a65f-1211-4471-a693-1841adb8ac62
                Copyright © 2020 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 12
                Funding
                Funded by: HHS | NIH | National Cancer Institute (NCI) 100000054
                Award ID: CA177322
                Award Recipient : Tariq M. Rana
                Categories
                Biological Sciences
                Medical Sciences

                m6a rna modification,melanoma,pd-1 blockade,immunotherapy enhancers,gvax

                Comments

                Comment on this article