3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Human Cytomegalovirus UL7, miR-US5-1, and miR-UL112-3p Inactivation of FOXO3a Protects CD34 + Hematopoietic Progenitor Cells from Apoptosis

      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

          Human cytomegalovirus (HCMV) causes serious disease in immunocompromised individuals and is a significant problem during transplantation. The virus can establish a latent infection in CD34 + hematopoietic progenitor cells (HPCs) and periodically reactivate to cause disease in the absence of an intact immune system.

          ABSTRACT

          Human cytomegalovirus (HCMV) infection of myeloid lineage cells, such as CD34 + hematopoietic progenitor cells (HPCs) or monocytes, results in the upregulation of antiapoptotic cellular proteins that protect the newly infected cells from programmed cell death. The mechanisms used by HCMV to regulate proapoptotic cellular proteins upon infection of CD34 + HPCs have not been fully explored. Here, we show that HCMV utilizes pUL7, a secreted protein that signals through the FLT3 receptor, and miR-US5-1 and miR-UL112-3p to reduce the abundance and activity of the proapoptotic transcription factor FOXO3a at early times after infection of CD34 + HPCs. Regulation of FOXO3a by pUL7, miR-US5-1, and miR-UL112 results in reduced expression of the proapoptotic BCL2L11 transcript and protection of CD34 + HPCs from virus-induced apoptosis. These data highlight the importance of both viral proteins and microRNAs (miRNAs) in protecting CD34 + HPCs from apoptosis at early times postinfection, allowing for the establishment of latency and maintenance of viral genome-containing cells.

          IMPORTANCE Human cytomegalovirus (HCMV) causes serious disease in immunocompromised individuals and is a significant problem during transplantation. The virus can establish a latent infection in CD34 + hematopoietic progenitor cells (HPCs) and periodically reactivate to cause disease in the absence of an intact immune system. What viral gene products are required for successful establishment of latency is still not fully understood. Here, we show that both a viral protein and viral miRNAs are required to prevent apoptosis after infection of CD34 + HPCs. HCMV pUL7 and miRNAs miR-US5-1 and miR-UL112-3p act to limit the expression and activation of the transcription factor FOXO3a, which in turn reduces expression of proapoptotic gene BCL2L11 and prevents virus-induced apoptosis in CD34 + HPCs.

          Related collections

          Most cited references56

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

          Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor.

          Survival factors can suppress apoptosis in a transcription-independent manner by activating the serine/ threonine kinase Akt, which then phosphorylates and inactivates components of the apoptotic machinery, including BAD and Caspase 9. In this study, we demonstrate that Akt also regulates the activity of FKHRL1, a member of the Forkhead family of transcription factors. In the presence of survival factors, Akt phosphorylates FKHRL1, leading to FKHRL1's association with 14-3-3 proteins and FKHRL1's retention in the cytoplasm. Survival factor withdrawal leads to FKHRL1 dephosphorylation, nuclear translocation, and target gene activation. Within the nucleus, FKHRL1 triggers apoptosis most likely by inducing the expression of genes that are critical for cell death, such as the Fas ligand gene.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            FoxO transcription factors; Regulation by AKT and 14-3-3 proteins.

            The forkhead box O (FoxO) transcription factor family is a key player in an evolutionary conserved pathway downstream of insulin and insulin-like growth factor receptors. The mammalian FoxO family consists of FoxO1, 3, 4 and 6, which share high similarity in their structure, function and regulation. FoxO proteins are involved in diverse cellular and physiological processes including cell proliferation, apoptosis, reactive oxygen species (ROS) response, longevity, cancer and regulation of cell cycle and metabolism. The regulation of FoxO protein function involves an intricate network of posttranslational modifications and protein-protein interactions that provide integrated cellular response to changing physiological conditions and cues. AKT was identified in early genetic and biochemical studies as a main regulator of FoxO function in diverse organisms. Though other FoxO regulatory pathways and mechanisms have been delineated since, AKT remains a key regulator of the pathway. The present review summarizes the current knowledge of FoxO regulation by AKT and 14-3-3 proteins, focusing on its mechanistic and structural aspects and discusses its crosstalk with the other FoxO regulatory mechanisms. This article is part of a Special Issue entitled: PI3K-AKT-FoxO axis in cancer and aging. 2011 Elsevier B.V. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation.

              The RAS-ERK pathway is known to play a pivotal role in differentiation, proliferation and tumour progression. Here, we show that Erk downregulates Forkhead box O 3a (FOXO3a) by directly interacting with and phosphorylating FOXO3a at Ser 294, Ser 344 and Ser 425, which consequently promotes cell proliferation and tumorigenesis. The ERK-phosphorylated FOXO3a degrades via an MDM2-mediated ubiquitin-proteasome pathway. However, the non-phosphorylated FOXO3a mutant is resistant to the interaction and degradation by murine double minute 2 (MDM2), thereby resulting in a strong inhibition of cell proliferation and tumorigenicity. Taken together, our study elucidates a novel pathway in cell growth and tumorigenesis through negative regulation of FOXO3a by RAS-ERK and MDM2.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mSphere
                mSphere
                msph
                msph
                mSphere
                mSphere
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2379-5042
                6 January 2021
                Jan-Feb 2021
                : 6
                : 1
                : e00986-20
                Affiliations
                [a ]Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, USA
                University of North Carolina, Chapel Hill
                Author notes
                Address correspondence to Patrizia Caposio, caposiop@ 123456ohsu.edu .

                Citation Hancock MH, Crawford LB, Perez W, Struthers HM, Mitchell J, Caposio P. 2021. Human cytomegalovirus UL7, miR-US5-1, and miR-UL112-3p inactivation of FOXO3a protects CD34 + hematopoietic progenitor cells from apoptosis. mSphere 6:e00986-20. https://doi.org/10.1128/mSphere.00986-20.

                Author information
                https://orcid.org/0000-0003-2945-0147
                https://orcid.org/0000-0003-1248-253X
                https://orcid.org/0000-0001-7579-849X
                Article
                mSphere00986-20
                10.1128/mSphere.00986-20
                7845589
                33408225
                9e04df99-21ac-480e-8d85-be30ed716d6a
                Copyright © 2021 Hancock et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 28 September 2020
                : 11 December 2020
                Page count
                supplementary-material: 4, Figures: 5, Tables: 0, Equations: 0, References: 55, Pages: 15, Words: 9242
                Funding
                Funded by: HHS | National Institutes of Health (NIH), https://doi.org/10.13039/100000002;
                Award ID: P01 AI127335
                Award Recipient :
                Funded by: HHS | National Institutes of Health (NIH), https://doi.org/10.13039/100000002;
                Award ID: P01 AI127335
                Award ID: R37 AI21640
                Award Recipient :
                Categories
                Research Article
                Host-Microbe Biology
                Custom metadata
                January/February 2021

                foxo3a,apoptosis,hematopoietic progenitor cells,human cytomegalovirus

                Comments

                Comment on this article