25
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Renal medullary carcinomas depend upon SMARCB1 loss and are sensitive to proteasome inhibition

      research-article
      1 , 2 , 3 , 3 , 3 , 2 , 2 , 3 , 3 , 2 , 3 , 2 , 2 , 3 , 2 , 3 , 3 , 3 , 1 , 2 , 4 , 2 , 3 , 3 , 5 , 1 , 2 , 1 , 1 , 2 , 3 , 1 , 2 , 4 , 4 , 2 , 1 , 2 , 3 , 3 , 3 , 3 , 2 , 1 , 2 , 1 , 2 , 6 , 2 , 3 , 2 , 3 , 7 , 2 , 3 , 7 , 3 , 2 , 3 , 7 ,
      ,
      eLife
      eLife Sciences Publications, Ltd
      renal medullary carcinoma, SMARCB1, MLN2238, ubiquitin-proteasome system, cell cycle, Human

      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

          Renal medullary carcinoma (RMC) is a rare and deadly kidney cancer in patients of African descent with sickle cell trait. We have developed faithful patient-derived RMC models and using whole-genome sequencing, we identified loss-of-function intronic fusion events in one SMARCB1 allele with concurrent loss of the other allele. Biochemical and functional characterization of these models revealed that RMC requires the loss of SMARCB1 for survival. Through integration of RNAi and CRISPR-Cas9 loss-of-function genetic screens and a small-molecule screen, we found that the ubiquitin-proteasome system (UPS) was essential in RMC. Inhibition of the UPS caused a G2/M arrest due to constitutive accumulation of cyclin B1. These observations extend across cancers that harbor SMARCB1 loss, which also require expression of the E2 ubiquitin-conjugating enzyme, UBE2C. Our studies identify a synthetic lethal relationship between SMARCB1-deficient cancers and reliance on the UPS which provides the foundation for a mechanism-informed clinical trial with proteasome inhibitors.

          eLife digest

          Renal medullary carcinoma (RMC for short) is a rare type of kidney cancer that affects teenagers and young adults. These patients are usually of African descent and carry one of the two genetic changes that cause sickle cell anemia. RMC is an aggressive disease without effective treatments and patients survive, on average, for only six to eight months after their diagnosis.

          Recent genetic studies found that most RMC cells have mutations that prevent them from producing a protein called SMARCB1. SMARCB1 normally acts as a so-called tumor suppressor, preventing cells from becoming cancerous. However, it was not clear whether RMCs always have to lose SMARCB1 if they are to survive and grow.

          Often, diseases are studied using laboratory-grown cells and tissues that have certain features of the disease. No such models had been created for RMC, which has slowed efforts to understand how the disease develops and find new treatments for it. Hong et al. therefore worked with patients to develop new lines of cells that can be used to study RMC in the laboratory. These RMC cells started dying when they were given copies of the SMARCB1 gene, which supports the theory that RMCs have to lose SMARCB1 in order to grow.

          Hong et al. then used a set of genetic reagents that can suppress or delete genes that are targeted by drugs, and followed this by testing a range of drugs on the RMC cells. Drugs and genetic reagents that reduced the activity of the proteasome – the structure inside cells that gets rid of old or unwanted proteins – caused the RMC cells to die. These proteasome inhibitor drugs also killed other kinds of cancer cells with SMARCB1 mutations.

          Proteasome inhibitors are already used to treat different types of cancer. Potentially, a clinical trial could be run to see if they will treat patients whose cancers lack SMARCB1. Further work is also needed to determine the exact link between SMARCB1 and the proteasome.

          Related collections

          Most cited references54

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

          Creation of human tumour cells with defined genetic elements.

          During malignant transformation, cancer cells acquire genetic mutations that override the normal mechanisms controlling cellular proliferation. Primary rodent cells are efficiently converted into tumorigenic cells by the coexpression of cooperating oncogenes. However, similar experiments with human cells have consistently failed to yield tumorigenic transformants, indicating a fundamental difference in the biology of human and rodent cells. The few reported successes in the creation of human tumour cells have depended on the use of chemical or physical agents to achieve immortalization, the selection of rare, spontaneously arising immortalized cells, or the use of an entire viral genome. We show here that the ectopic expression of the telomerase catalytic subunit (hTERT) in combination with two oncogenes (the simian virus 40 large-T oncoprotein and an oncogenic allele of H-ras) results in direct tumorigenic conversion of normal human epithelial and fibroblast cells. These results demonstrate that disruption of the intracellular pathways regulated by large-T, oncogenic ras and telomerase suffices to create a human tumor cell.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Correlating chemical sensitivity and basal gene expression reveals mechanism of action

            Changes in cellular gene expression in response to small-molecule or genetic perturbations have yielded signatures that can connect unknown mechanisms of action (MoA) to ones previously established. We hypothesized that differential basal gene expression could be correlated with patterns of small-molecule sensitivity across many cell lines to illuminate the actions of compounds whose MoA are unknown. To test this idea, we correlated the sensitivity patterns of 481 compounds with ~19,000 basal transcript levels across 823 different human cancer cell lines and identified selective outlier transcripts. This process yielded many novel mechanistic insights, including the identification of activation mechanisms, cellular transporters, and direct protein targets. We found that ML239, originally identified in a phenotypic screen for selective cytotoxicity in breast cancer stem-like cells, most likely acts through activation of fatty acid desaturase 2 (FADS2). These data and analytical tools are available to the research community through the Cancer Therapeutics Response Portal.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset.

              Identifying genetic alterations that prime a cancer cell to respond to a particular therapeutic agent can facilitate the development of precision cancer medicines. Cancer cell-line (CCL) profiling of small-molecule sensitivity has emerged as an unbiased method to assess the relationships between genetic or cellular features of CCLs and small-molecule response. Here, we developed annotated cluster multidimensional enrichment analysis to explore the associations between groups of small molecules and groups of CCLs in a new, quantitative sensitivity dataset. This analysis reveals insights into small-molecule mechanisms of action, and genomic features that associate with CCL response to small-molecule treatment. We are able to recapitulate known relationships between FDA-approved therapies and cancer dependencies and to uncover new relationships, including for KRAS-mutant cancers and neuroblastoma. To enable the cancer community to explore these data, and to generate novel hypotheses, we created an updated version of the Cancer Therapeutic Response Portal (CTRP v2).
                Bookmark

                Author and article information

                Contributors
                Role: Senior Editor
                Role: Reviewing Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                12 March 2019
                2019
                : 8
                : e44161
                Affiliations
                [1 ]Boston Children’s Hospital BostonUnited States
                [2 ]Dana-Farber Cancer Institute BostonUnited States
                [3 ]Broad Institute of Harvard and MIT CambridgeUnited States
                [4 ]Rare Cancer Research Foundation DurhamUnited States
                [5 ]RMC Support North CharlestonUnited States
                [6 ]St. Jude Children’s Research Hospital MemphisUnited States
                [7 ]Brigham and Women’s Hospital BostonUnited States
                Calico Life Sciences United States
                Memorial Sloan Kettering Cancer Center United States
                Memorial Sloan Kettering Cancer Center United States
                Memorial Sloan Kettering Cancer Center United States
                UT Southwestern Medical Center United States
                Author information
                http://orcid.org/0000-0003-0374-1667
                http://orcid.org/0000-0001-6303-3609
                http://orcid.org/0000-0002-6795-6336
                http://orcid.org/0000-0003-2840-9791
                Article
                44161
                10.7554/eLife.44161
                6436895
                30860482
                45480362-e6bf-4a50-8838-4471070414c0
                © 2019, Hong et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 05 December 2018
                : 03 March 2019
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100001445, Alex's Lemonade Stand Foundation for Childhood Cancer;
                Award ID: Young Investigator Award
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000043, American Association for Cancer Research;
                Award ID: 14-40-31-HONG
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000048, American Cancer Society;
                Award ID: 132943-MRSG-18-202-01-TBG
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100006823, Boston Children's Hospital;
                Award ID: OFD BTREC CDA
                Award Recipient :
                Funded by: Cure AT/RT;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100006315, CureSearch for Children's Cancer;
                Award ID: 328545
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100007886, Dana-Farber Cancer Institute;
                Award ID: Wong Family Award
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100009633, Eunice Kennedy Shriver National Institute of Child Health and Human Development;
                Award ID: K12 HD052896
                Award Recipient :
                Funded by: Katie Moore Foundation;
                Award Recipient :
                Funded by: Merkin Family Foundation;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000054, National Cancer Institute;
                Award ID: U01 CA176058
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000054, National Cancer Institute;
                Award ID: U01 CA217848
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000054, National Cancer Institute;
                Award ID: P50 CA101942
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000057, National Institute of General Medical Sciences;
                Award ID: T32 GM007753
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000057, National Institute of General Medical Sciences;
                Award ID: T32 GM007226
                Award Recipient :
                Funded by: Team Path to Cure;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000005, U.S. Department of Defense;
                Award ID: W81XWH-15-1-0659
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cancer Biology
                Human Biology and Medicine
                Custom metadata
                Faithful models of RMC require SMARCB1 loss for survival, and genetic and small-molecule screens identify inhibition of the ubiquitin-proteasome system (UPS) as a potential therapeutic approach for SMARCB1 deficient cancers.

                Life sciences
                renal medullary carcinoma,smarcb1,mln2238,ubiquitin-proteasome system,cell cycle,human
                Life sciences
                renal medullary carcinoma, smarcb1, mln2238, ubiquitin-proteasome system, cell cycle, human

                Comments

                Comment on this article