8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Discovery and Targeting of the Signaling Controls of PNPLA3 to Effectively Reduce Transcription, Expression, and Function in Pre-Clinical NAFLD/NASH Settings

      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.

          Abstract

          Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are emerging worldwide epidemics, projected to become the leading cause of liver transplants. The strongest genetic risk factor for NAFLD/NASH susceptibility and progression is a single-nucleotide polymorphism (SNP) in the patatin-like phospholipase domain-containing 3 gene ( PNPLA3), rs738409, encoding the missense mutation I148M. This aminoacidic substitution interferes with the normal remodeling of lipid droplets in hepatocytes. It is also thought to play a key role in promoting liver fibrosis by inhibiting the release of retinol from hepatic stellate cells. Reducing PNPLA3 levels in individuals homozygous for 148M may be an effective treatment for the entire spectrum of NAFLD, based on gene dosage analysis in the human population, as well as the protective effect of another naturally occurring SNP (rs2294918) in PNPLA3 which, when co-inherited, reduces PNPLA3 mRNA levels to 50% and counteracts disease risk. By screening a clinical compound library targeting specific signaling pathways active in primary human hepatocytes, we identified momelotinib, a drug evaluated in clinical trials to treat myelofibrosis, as a potent down-regulator of PNPLA3 expression, across all genotypes. We found that momelotinib treatment yielded >80% reduction in PNPLA3 mRNA in human primary hepatocytes and stellate cells, as well as in vivo via acute and chronic treatment of WT mice. Using a human multilineage 3D spheroid model of NASH homozygous for the PNPLA3 mutant protein, we additionally show that it decreases PNPLA3 mRNA as well as intracellular lipid content. Furthermore, we show that the effects on PNPLA3 coincide with changes in chromatin accessibility within regulatory regions of the PNPLA3 locus, consistent with inhibition occurring at the level of transcription. In addition to its primary reported targets, the JAK kinases, momelotinib inhibits several non-JAK kinases, including ACVR1. Using a combination of targeted siRNA knockdowns and signaling pathway perturbations, we show that momelotinib reduces the expression of the PNPLA3 gene largely through the inhibition of BMP signaling rather than the JAK/STAT pathway. Overall, our work identified momelotinib as a potential NASH therapeutic and uncovered previously unrecognized connections between signaling pathways and PNPLA3. These pathways may be exploited by drug modalities to “tune down” the level of gene expression, and therefore offer a potential therapeutic benefit to a high at-risk subset of NAFLD/NASH patients.

          Related collections

          Most cited references29

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

          Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease

          Nonalcoholic fatty liver disease (NAFLD) is a burgeoning health problem of unknown etiology that varies in prevalence among ethnic groups. To identify genetic variants contributing to differences in hepatic fat content, we performed a genome-wide association scan of nonsynonymous sequence variations (n=9,229) in a multiethnic population. An allele in PNPLA3 (rs738409; I148M) was strongly associated with increased hepatic fat levels (P=5.9×10−10) and with hepatic inflammation (P=3.7×10−4). The allele was most common in Hispanics, the group most susceptible to NAFLD; hepatic fat content was > 2-fold higher in PNPLA3-148M homozygotes than in noncarriers. Resequencing revealed another allele associated with lower hepatic fat content in African-Americans, the group at lowest risk of NAFLD. Thus, variation in PNPLA3 contributes to ethnic and inter-individual differences in hepatic fat content and susceptibility to NAFLD.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues

            The Omni-ATAC protocol improves the signal-to-background ratio in chromatin accessibility profiles and is suitable for a range of cell lines and primary cell types, as well as frozen tissue.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Organization and function of the 3D genome

              Understanding how chromatin is organized within the nucleus and how this 3D architecture influences gene regulation, cell fate decisions and evolution are major questions in cell biology. Despite spectacular progress in this field, we still know remarkably little about the mechanisms underlying chromatin structure and how it can be established, reset and maintained. In this Review, we discuss the insights into chromatin architecture that have been gained through recent technological developments in quantitative biology, genomics and cell and molecular biology approaches and explain how these new concepts have been used to address important biological questions in development and disease.
                Bookmark

                Author and article information

                Journal
                Cells
                Cells
                cells
                Cells
                MDPI
                2073-4409
                07 October 2020
                October 2020
                : 9
                : 10
                : 2247
                Affiliations
                [1 ]CAMP4 Therapeutics, Cambridge, MA 02139, USA; vaishnavi.rajagopal@ 123456gmail.com (V.R.); smith@ 123456camp4tx.com (C.S.); cohick@ 123456camp4tx.com (E.C.); whissell@ 123456camp4tx.com (G.W.); gamboa@ 123456camp4tx.com (M.G.); pai@ 123456camp4tx.com (R.P.); sigova@ 123456camp4tx.com (A.S.); iris@ 123456grossmail.com (I.G.); bumcrot@ 123456camp4tx.com (D.B.); sehgal@ 123456camp4tx.com (A.S.)
                [2 ]Department of Molecular and Clinical Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Kavitha.Sasidharan@ 123456wlab.gu.se (K.S.); stefano.romeo@ 123456wlab.gu.se (S.R.)
                Author notes
                [* ]Correspondence: schwartz@ 123456camp4tx.com (B.E.S.); Piero.Pingitore@ 123456wlab.gu.se (P.P.); Tel.: +1-617-651-8867 (B.E.S.)
                Author information
                https://orcid.org/0000-0002-0824-9148
                https://orcid.org/0000-0003-3912-7458
                https://orcid.org/0000-0002-2478-225X
                Article
                cells-09-02247
                10.3390/cells9102247
                7600576
                33036387
                15b35fb1-c911-4f31-ad23-3bb2851b0826
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 16 July 2020
                : 05 October 2020
                Categories
                Article

                nash,nafld,pnpla3,momelotinib,cyt-387,acvr1,stellate,jak,bmp signaling,fibrosis,rs2294918

                Comments

                Comment on this article