1
|
Bastawecy IM, Abdelmonem M, Afify AF, Saad N, Shirosaki Y, Abdullah CAC, El Naggar RF, Rohaim MA, Munir M. Viral contamination in cell culture: analyzing the impact of Epstein Barr virus and Ovine Herpesvirus 2. Front Microbiol 2025; 16:1442321. [PMID: 40071201 PMCID: PMC11893573 DOI: 10.3389/fmicb.2025.1442321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Cell culture techniques are increasingly favored over animal models due to rising costs, time constraints, and ethical concerns regarding animal use. These techniques serve critical roles in disease modeling, drug screening, drug discovery, and toxicity analysis. Notably, cell cultures facilitate primary virus isolation, infectivity assays, biochemical studies, and vaccine production. However, viral contamination in cell cultures poses significant challenges, particularly due to the necessity for complex and sophisticated detection methods. Among the prevalent viruses, Epstein Barr virus (EBV) is ubiquitous across human populations, infecting approximately 98% of individuals. Despite its prevalence, the detection of EBV is often not considered a safety priority, as its detection methods are well-established, including PCR assays that can identify both active and latent forms of the virus. Conversely, ovine herpesvirus 2 (OvHV-2), a relative of EBV, presents a critical concern due to its ability to infect a wide range of organs and species, including over 33 animal species and nearly all domestic sheep. This makes the detection of OvHV-2 crucial for the safety of cell cultures across various species. The literature reveals a gap in the comprehensive understanding of both EBV and OvHv-2 detection in cell culture systems, highlighting an urgent need for developing robust detection methodologies specific to EBV and OvHv-2 to ensure bioprocess safety.
Collapse
Affiliation(s)
- Iman M. Bastawecy
- Department of Virology, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Mohamed Abdelmonem
- Department of Physics, Faculty of Science, Universiti Putra Malaysia (UPM), Selangor Darul Ehsan, Malaysia
| | - Ahmed F. Afify
- Department of Virology, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Norazalina Saad
- Laboratory of Cancer Research UPM-MAKNA (CANRES), Institute of Bioscience, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| | - Yuki Shirosaki
- Faculty of Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
- Research Center of Synthetic Biology, Kyushu Institute of Technology, Kitakyushu, Japan
- Collaborative Research Centre for Green Materials on Environmental Technology, Kyushu Institute of Technology, Kitakyushu, Japan
| | | | - Rania F. El Naggar
- Department of Virology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Mohammed A. Rohaim
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
2
|
Anisenko AN, Nefedova AA, Kireev II, Gottikh MB. Post-Integrational DNA Repair of HIV-1 Is Associated with Activation of the DNA-PK and ATM Cellular Protein Kinases and Phosphorylation of Their Targets. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1122-1132. [PMID: 38981705 DOI: 10.1134/s0006297924060117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 07/11/2024]
Abstract
Integration of the DNA copy of HIV-1 genome into the cellular genome results in series of damages, repair of which is critical for successful replication of the virus. We have previously demonstrated that the ATM and DNA-PK kinases, normally responsible for repairing double-strand breaks in the cellular DNA, are required to initiate the HIV-1 DNA postintegrational repair, even though integration does not result in DNA double-strand breaks. In this study, we analyzed changes in phosphorylation status of ATM (pSer1981), DNA-PK (pSer2056), and their related kinase ATR (pSer428), as well as their targets: Chk1 (pSer345), Chk2 (pThr68), H2AX (pSer139), and p53 (pSer15) during the HIV-1 DNA postintegrational repair. We have shown that ATM and DNA-PK, but not ATR, undergo autophosphorylation during postintegrational DNA repair and phosphorylate their target proteins Chk2 and H2AX. These data indicate common signaling mechanisms between the double-strand DNA break repair and postintegrational repair of HIV-1 DNA.
Collapse
Affiliation(s)
- Andrey N Anisenko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Anastasiia A Nefedova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Igor I Kireev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Marina B Gottikh
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
3
|
Li S, Liu B, Tan M, Juillard F, Szymula A, Álvarez Á, Van Sciver N, George A, Ramachandran A, Raina K, Tumuluri VS, Costa C, Simas J, Kaye K. Kaposi's sarcoma herpesvirus exploits the DNA damage response to circularize its genome. Nucleic Acids Res 2024; 52:1814-1829. [PMID: 38180827 PMCID: PMC10899755 DOI: 10.1093/nar/gkad1224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/05/2023] [Accepted: 12/16/2023] [Indexed: 01/07/2024] Open
Abstract
To establish lifelong, latent infection, herpesviruses circularize their linear, double-stranded, DNA genomes through an unknown mechanism. Kaposi's sarcoma (KS) herpesvirus (KSHV), a gamma herpesvirus, is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman's disease. KSHV persists in latently infected cells as a multi-copy, extrachromosomal episome. Here, we show the KSHV genome rapidly circularizes following infection, and viral protein expression is unnecessary for this process. The DNA damage response (DDR) kinases, ATM and DNA-PKcs, each exert roles, and absence of both severely compromises circularization and latency. These deficiencies were rescued by expression of ATM and DNA-PKcs, but not catalytically inactive mutants. In contrast, γH2AX did not function in KSHV circularization. The linear viral genomic ends resemble a DNA double strand break, and non-homologous DNA end joining (NHEJ) and homologous recombination (HR) reporters indicate both NHEJ and HR contribute to KSHV circularization. Last, we show, similar to KSHV, ATM and DNA-PKcs have roles in circularization of the alpha herpesvirus, herpes simplex virus-1 (HSV-1), while γH2AX does not. Therefore, the DDR mediates KSHV and HSV-1 circularization. This strategy may serve as a general herpesvirus mechanism to initiate latency, and its disruption may provide new opportunities for prevention of herpesvirus disease.
Collapse
Affiliation(s)
- Shijun Li
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Bing Liu
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Min Tan
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Franceline Juillard
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Agnieszka Szymula
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Ángel L Álvarez
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Van Sciver
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Athira George
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Akshaya Ramachandran
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Komal Raina
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Vinayak Sadasivam Tumuluri
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Catarina N Costa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research, Palma de Cima, 1649-023 Lisboa, Portugal
| | - J Pedro Simas
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Kenneth M Kaye
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
4
|
Weizel I, Shavit T, Shuli Y, Adler Lazarovich C, Halevi R, Ben Ari T, Yaacobi-Artzi S, Bentov Y, Feldman B, Hershko Klement A. Outcomes of BRCA pre-implantation genetic testing according to the parental mutation origin: a cohort study. Reprod Biol Endocrinol 2024; 22:8. [PMID: 38172815 PMCID: PMC10762833 DOI: 10.1186/s12958-023-01180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The process of gamete formation and early embryonic development involves rapid DNA replication, chromosome segregation and cell division. These processes may be affected by mutations in the BRCA1/2 genes. The aim of this study was to evaluate BRCA mutation inheritance and its effect on early embryonic development according to the parental origin of the mutation. The study question was approached by analyzing in vitro fertilization cycles (IVF) that included pre-implantation testing (PGT-M) for a BRCA gene mutation. METHODS This retrospective cohort study compared cycles of pre-implantation genetic testing for mutations (PGT-M) between male and female patients diagnosed with BRCA 1/2 mutations (cases), to a control group of two other mutations with dominant inheritance (myotonic dystrophy (MD) and polycystic kidney disease (PKD)). Results were compared according to mutation type and through a generalized linear model analysis. RESULTS The cohort included 88 PGT-M cycles (47 BRCA and 41 non-BRCA) among 50 patients. Maternal and paternal ages at oocyte retrieval were comparable between groups. When tested per cycle, FSH dose, maximum estradiol level, oocytes retrieved, number of zygotes, and number of embryos available for biopsy and affected embryos, were not significantly different among mutation types. All together 444 embryos were biopsied: the rate of affected embryos was comparable between groups. Among BRCA patients, the proportion of affected embryos was similar between maternal and paternal mutation origin (p = 0.24). In a generalized linear model analysis, the relative oocyte yield in maternal BRCA patients was significantly lower (0.7, as related to the non BRCA group)(p < 0.001). Zygote formation and blastulation were not affected by the BRCA gene among paternal cases (P = 0.176 and P = 0.293 respectively), nor by paternal versus maternal BRCA carriage (P = 0.904 and P = 0.149, respectively). CONCLUSIONS BRCA PGT-M cycles performed similarly compared to non-BRCA cycles. Inheritance rate and cycle parameters were not affected by the parental origin of the mutation.
Collapse
Affiliation(s)
- Ilana Weizel
- The IVF Unit, Department of Obstetrics and Gynecology, Hadassah Mount Scopus- Hebrew University Medical Center, Mt Scopus, Jerusalem, 9112001, Israel
| | - Tal Shavit
- The IVF unit, Assuta Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Yulia Shuli
- The Unit for Medical Genetics, Assuta Medical Center, Tel Aviv, Israel
| | - Chana Adler Lazarovich
- The IVF Unit, Department of Obstetrics and Gynecology, Hadassah Mount Scopus- Hebrew University Medical Center, Mt Scopus, Jerusalem, 9112001, Israel
| | - Rivka Halevi
- The Unit for Medical Genetics, Assuta Medical Center, Tel Aviv, Israel
| | - Tal Ben Ari
- The IVF unit, Assuta Medical Center, Tel Aviv, Israel
| | | | - Yaakov Bentov
- The IVF Unit, Department of Obstetrics and Gynecology, Hadassah Mount Scopus- Hebrew University Medical Center, Mt Scopus, Jerusalem, 9112001, Israel
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Baruch Feldman
- The Unit for Medical Genetics, Assuta Medical Center, Tel Aviv, Israel
| | - Anat Hershko Klement
- The IVF Unit, Department of Obstetrics and Gynecology, Hadassah Mount Scopus- Hebrew University Medical Center, Mt Scopus, Jerusalem, 9112001, Israel.
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
5
|
Anisenko A, Galkin S, Mikhaylov AA, Khrenova MG, Agapkina Y, Korolev S, Garkul L, Shirokova V, Ikonnikova VA, Korlyukov A, Dorovatovskii P, Baranov M, Gottikh M. KuINins as a New Class of HIV-1 Inhibitors That Block Post-Integration DNA Repair. Int J Mol Sci 2023; 24:17354. [PMID: 38139188 PMCID: PMC10744174 DOI: 10.3390/ijms242417354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Integration of HIV-1 genomic cDNA results in the formation of single-strand breaks in cellular DNA, which must be repaired for efficient viral replication. Post-integration DNA repair mainly depends on the formation of the HIV-1 integrase complex with the Ku70 protein, which promotes DNA-PK assembly at sites of integration and its activation. Here, we have developed a first-class inhibitor of the integrase-Ku70 complex formation that inhibits HIV-1 replication in cell culture by acting at the stage of post-integration DNA repair. This inhibitor, named s17, does not affect the main cellular function of Ku70, namely its participation in the repair of double-strand DNA breaks through the non-homologous end-joining pathway. Using a molecular dynamics approach, we have constructed a model for the interaction of s17 with Ku70. According to this model, the interaction of two phenyl radicals of s17 with the L76 residue of Ku70 is important for this interaction. The requirement of two phenyl radicals in the structure of s17 for its inhibitory properties was confirmed using a set of s17 derivatives. We propose to stimulate compounds that inhibit post-integration repair by disrupting the integrase binding to Ku70 KuINins.
Collapse
Affiliation(s)
- Andrey Anisenko
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.G.K.); (Y.A.); (S.K.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (L.G.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Simon Galkin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (L.G.)
| | - Andrey A. Mikhaylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia (V.S.); (V.A.I.); (M.B.)
| | - Maria G. Khrenova
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.G.K.); (Y.A.); (S.K.)
- Federal Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia
| | - Yulia Agapkina
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.G.K.); (Y.A.); (S.K.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Sergey Korolev
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.G.K.); (Y.A.); (S.K.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Lidia Garkul
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (L.G.)
| | - Vasilissa Shirokova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia (V.S.); (V.A.I.); (M.B.)
- Higher Chemical College, D.I. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia
| | - Viktoria A. Ikonnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia (V.S.); (V.A.I.); (M.B.)
- Higher Chemical College, D.I. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia
| | - Alexander Korlyukov
- Nesmeyanov Institute of Organoelement Compounds, 119334 Moscow, Russia;
- Institute of Translational Medicine and Institute of Pharmacy and Medicinal Chemistry, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | | | - Mikhail Baranov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia (V.S.); (V.A.I.); (M.B.)
- Institute of Translational Medicine and Institute of Pharmacy and Medicinal Chemistry, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Marina Gottikh
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (L.G.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
6
|
Anisenko A, Nefedova A, Agapkina Y, Gottikh M. Both ATM and DNA-PK Are the Main Regulators of HIV-1 Post-Integrational DNA Repair. Int J Mol Sci 2023; 24:ijms24032797. [PMID: 36769109 PMCID: PMC9917498 DOI: 10.3390/ijms24032797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The integration of a DNA copy of an HIV-1 RNA genome into the host genome, carried out by the viral enzyme integrase, results in the formation of single-stranded gaps in cellular DNA that must be repaired. Here, we have analyzed the involvement of the PI3K kinases, ATM, ATR, and DNA-PKcs, which are important players in the DNA damage response (DDR) in HIV-1 post-integrational DNA repair (PIR). The participation of the DNA-PK complex in HIV-1 PIR has been previously shown, and the formation of a complex between the viral integrase and the DNA-PK subunit, Ku70, has been found to be crucial for efficient PIR. Now, we have shown that the inhibition of both DNA-PKcs and ATM, but not ATR, significantly reduces PIR efficiency. The activation of both kinases is a sequential process, where one kinase, being activated, activates the other, and it occurs simultaneously with the integration of viral DNA. This fact suggests that the activation of both kinases triggers PIR. Most interestingly, the activation of not only DNA-PKcs, but also ATM depends on the complex formation between integrase and Ku70. The elucidation of the interactions between viruses and DDR is important both for understanding the modulation of host cell functions by these pathogens and for developing new approaches to combat viral infections.
Collapse
Affiliation(s)
- Andrey Anisenko
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Anastasiia Nefedova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Yulia Agapkina
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marina Gottikh
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
7
|
Dutta B, Osato M. The RUNX Family, a Novel Multifaceted Guardian of the Genome. Cells 2023; 12:255. [PMID: 36672189 PMCID: PMC9856552 DOI: 10.3390/cells12020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The DNA repair machinery exists to protect cells from daily genetic insults by orchestrating multiple intrinsic and extrinsic factors. One such factor recently identified is the Runt-related transcription factor (RUNX) family, a group of proteins that act as a master transcriptional regulator for multiple biological functions such as embryonic development, stem cell behaviors, and oncogenesis. A significant number of studies in the past decades have delineated the involvement of RUNX proteins in DNA repair. Alterations in RUNX genes cause organ failure and predisposition to cancers, as seen in patients carrying mutations in the other well-established DNA repair genes. Herein, we review the currently existing findings and provide new insights into transcriptional and non-transcriptional multifaceted regulation of DNA repair by RUNX family proteins.
Collapse
Affiliation(s)
- Bibek Dutta
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Motomi Osato
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
8
|
Lopez A, Nichols Doyle R, Sandoval C, Nisson K, Yang V, Fregoso OI. Viral Modulation of the DNA Damage Response and Innate Immunity: Two Sides of the Same Coin. J Mol Biol 2022; 434:167327. [PMID: 34695379 PMCID: PMC9119581 DOI: 10.1016/j.jmb.2021.167327] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
The DDR consists of multiple pathways that sense, signal, and respond to anomalous DNA. To promote efficient replication, viruses have evolved to engage and even modulate the DDR. In this review, we will discuss a select set of diverse viruses and the range of mechanisms they evolved to interact with the DDR and some of the subsequent cellular consequences. There is a dichotomy in that the DDR can be both beneficial for viruses yet antiviral. We will also review the connection between the DDR and innate immunity. Previously believed to be disparate cellular functions, more recent research is emerging that links these processes. Furthermore, we will discuss some discrepancies in the literature that we propose can be remedied by utilizing more consistent DDR-focused assays. By doing so, we hope to obtain a much clearer understanding of how broadly these mechanisms and phenotypes are conserved among all viruses. This is crucial for human health since understanding how viruses manipulate the DDR presents an important and tractable target for antiviral therapies.
Collapse
Affiliation(s)
- Andrew Lopez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Randilea Nichols Doyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Carina Sandoval
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Karly Nisson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Vivian Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Oliver I Fregoso
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Liu B, Wang K, Wu J, Hu Y, Yang X, Xu L, Sun W, Jia X, Wu J, Fu S, Qiao Y, Zhang X. Association of APEX1 and XRCC1 Gene Polymorphisms With HIV-1 Infection Susceptibility and AIDS Progression in a Northern Chinese MSM Population. Front Genet 2022; 13:861355. [PMID: 35368687 PMCID: PMC8966225 DOI: 10.3389/fgene.2022.861355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/21/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Some studies have shown that the base excision repair (BER) pathway has an effect on HIV-1 replication. APEX1 and XRCC1 as key BER genes may affect DNA repair capacity. However, the roles of single nucleotide polymorphisms (SNPs) in APEX1 and XRCC1 and their impact on HIV-1 infection and AIDS progression remain unclear. Methods: A custom-designed 48-Plex SNPscan Kit was used for detection of single nucleotide polymorphisms. 601 HIV-1-infected men who have sex with men (MSM) and 624 age-matched healthy individuals were recruited in northern China. Four SNPs (rs1130409, rs1760944, rs2307486 and rs3136817) in APEX1 gene and three SNPs (rs1001581, rs25487 and rs25489) in XRCC1 gene were genotyped. The generalized multifactor dimension reduction (GMDR) method was used to identify the SNP-SNP interactions. Results: In this study, rs1130409 G allele, rs1001581 C allele and rs25487 C allele were associated with a higher risk of HIV-1 infection susceptibility (p = 0.020, p = 0.007 and p = 0.032, respectively). The frequencies of APEX1 haplotype TT and XRCC1 haplotype CT showed significant differences between cases and controls (p = 0.0372 and p = 0.0189, respectively). Interestingly, stratified analysis showed that the frequency of rs1001581 C allele was significantly higher in AIDS patients with the CD4+ T-lymphocyte count <200 cells/μl than those with >200 cells/μl (p = 0.022). Moreover, significant gene-gene interactions among rs1130409, rs1001581 and rs25487 were identified by GMDR (p = 0.0107). Specially, individuals with five to six risk alleles have a higher susceptibility to HIV-1 infection than those with zero to two risk alleles (p < 0.001). Conclusion:APEX1 and XRCC1 gene polymorphisms were associated with the susceptibility to HIV-1 infection and AIDS progression in MSM populations in northern China.
Collapse
Affiliation(s)
- Bangquan Liu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Kaili Wang
- The Second Hospital of Heilongjiang Province, Harbin, China
| | - Jiawei Wu
- College of Basic Medicine, Harbin Medical University-Daqing, Daqing, China
| | - Yuanting Hu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Xun Yang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lidan Xu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Wenjing Sun
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Xueyuan Jia
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jie Wu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Songbin Fu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Yuandong Qiao
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
- *Correspondence: Yuandong Qiao, ; Xuelong Zhang,
| | - Xuelong Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
- *Correspondence: Yuandong Qiao, ; Xuelong Zhang,
| |
Collapse
|
10
|
Marty FH, Bettamin L, Thouard A, Bourgade K, Allart S, Larrieu G, Malnou CE, Gonzalez-Dunia D, Suberbielle E. Borna disease virus docks on neuronal DNA double-strand breaks to replicate and dampens neuronal activity. iScience 2022; 25:103621. [PMID: 35024577 PMCID: PMC8724971 DOI: 10.1016/j.isci.2021.103621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/11/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022] Open
Abstract
Borna disease viruses (BoDV) have recently emerged as zoonotic neurotropic pathogens. These persistent RNA viruses assemble nuclear replication centers (vSPOT) in close interaction with the host chromatin. However, the topology of this interaction and its consequences on neuronal function remain unexplored. In neurons, DNA double-strand breaks (DSB) have been identified as novel epigenetic mechanisms regulating neurotransmission and cognition. Activity-dependent DSB contribute critically to neuronal plasticity processes, which could be impaired upon infection. Here, we show that BoDV-1 infection, or the singled-out expression of viral Nucleoprotein and Phosphoprotein, increases neuronal DSB levels. Of interest, inducing DSB promoted the recruitment anew of vSPOT colocalized with DSB and increased viral RNA replication. BoDV-1 persistence decreased neuronal activity and response to stimulation by dampening the surface expression of glutamate receptors. Taken together, our results propose an original mechanistic cross talk between persistence of an RNA virus and neuronal function, through the control of DSB levels. BoDV-1, its Nucleoprotein or Phosphoprotein cause neuronal DNA double-strand breaks (DSB) DNA double-strand breaks co-localize with BoDV-1 replication factories DNA DSB recruits BoDV-1 replication factories and promotes viral replication BoDV-1 inhibits neuronal activity by impeding surface expression of GluN2A receptors
Collapse
Affiliation(s)
| | - Luca Bettamin
- Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
- LAAS-CNRS, Toulouse, France
| | - Anne Thouard
- Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Karine Bourgade
- Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Sophie Allart
- Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | | | | | | | - Elsa Suberbielle
- Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
- Corresponding author
| |
Collapse
|
11
|
Passos DO, Li M, Craigie R, Lyumkis D. Retroviral integrase: Structure, mechanism, and inhibition. Enzymes 2021; 50:249-300. [PMID: 34861940 DOI: 10.1016/bs.enz.2021.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The retroviral protein Integrase (IN) catalyzes concerted integration of viral DNA into host chromatin to establish a permanent infection in the target cell. We learned a great deal about the mechanism of catalytic integration through structure/function studies over the previous four decades of IN research. As one of three essential retroviral enzymes, IN has also been targeted by antiretroviral drugs to treat HIV-infected individuals. Inhibitors blocking the catalytic integration reaction are now state-of-the-art drugs within the antiretroviral therapy toolkit. HIV-1 IN also performs intriguing non-catalytic functions that are relevant to the late stages of the viral replication cycle, yet this aspect remains poorly understood. There are also novel allosteric inhibitors targeting non-enzymatic functions of IN that induce a block in the late stages of the viral replication cycle. In this chapter, we will discuss the function, structure, and inhibition of retroviral IN proteins, highlighting remaining challenges and outstanding questions.
Collapse
Affiliation(s)
| | - Min Li
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - Robert Craigie
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - Dmitry Lyumkis
- The Salk Institute for Biological Studies, La Jolla, CA, United States; The Scripps Research Institute, La Jolla, CA, United States.
| |
Collapse
|
12
|
Hayman TJ, Glazer PM. Regulation of the Cell-Intrinsic DNA Damage Response by the Innate Immune Machinery. Int J Mol Sci 2021; 22:12761. [PMID: 34884568 PMCID: PMC8657976 DOI: 10.3390/ijms222312761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
Maintenance of genomic integrity is crucial for cell survival. As such, elegant DNA damage response (DDR) systems have evolved to ensure proper repair of DNA double-strand breaks (DSBs) and other lesions that threaten genomic integrity. Towards this end, most therapeutic studies have focused on understanding of the canonical DNA DSB repair pathways to enhance the efficacy of DNA-damaging therapies. While these approaches have been fruitful, there has been relatively limited success to date and potential for significant normal tissue toxicity. With the advent of novel immunotherapies, there has been interest in understanding the interactions of radiation therapy with the innate and adaptive immune responses, with the ultimate goal of enhancing treatment efficacy. While a substantial body of work has demonstrated control of the immune-mediated (extrinsic) responses to DNA-damaging therapies by several innate immune pathways (e.g., cGAS-STING and RIG-I), emerging work demonstrates an underappreciated role of the innate immune machinery in directly regulating tumor cell-intrinsic/cell-autonomous responses to DNA damage.
Collapse
Affiliation(s)
- Thomas J. Hayman
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Peter M. Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA;
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
13
|
Predicted Cellular Interactors of the Endogenous Retrovirus-K Integrase Enzyme. Microorganisms 2021; 9:microorganisms9071509. [PMID: 34361946 PMCID: PMC8303831 DOI: 10.3390/microorganisms9071509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022] Open
Abstract
Integrase (IN) enzymes are found in all retroviruses and are crucial in the retroviral integration process. Many studies have revealed how exogenous IN enzymes, such as the human immunodeficiency virus (HIV) IN, contribute to altered cellular function. However, the same consideration has not been given to viral IN originating from symbionts within our own DNA. Endogenous retrovirus-K (ERVK) is pathologically associated with neurological and inflammatory diseases along with several cancers. The ERVK IN interactome is unknown, and the question of how conserved the ERVK IN protein-protein interaction motifs are as compared to other retroviral integrases is addressed in this paper. The ERVK IN protein sequence was analyzed using the Eukaryotic Linear Motif (ELM) database, and the results are compared to ELMs of other betaretroviral INs and similar eukaryotic INs. A list of putative ERVK IN cellular protein interactors was curated from the ELM list and submitted for STRING analysis to generate an ERVK IN interactome. KEGG analysis was used to identify key pathways potentially influenced by ERVK IN. It was determined that the ERVK IN potentially interacts with cellular proteins involved in the DNA damage response (DDR), cell cycle, immunity, inflammation, cell signaling, selective autophagy, and intracellular trafficking. The most prominent pathway identified was viral carcinogenesis, in addition to select cancers, neurological diseases, and diabetic complications. This potentiates the role of ERVK IN in these pathologies via protein-protein interactions facilitating alterations in key disease pathways.
Collapse
|
14
|
Guo G, Gao M, Gao X, Zhu B, Huang J, Tu X, Kim W, Zhao F, Zhou Q, Zhu S, Wu Z, Yan Y, Zhang Y, Zeng X, Zhu Q, Yin P, Luo K, Sun J, Deng M, Lou Z. Reciprocal regulation of RIG-I and XRCC4 connects DNA repair with RIG-I immune signaling. Nat Commun 2021; 12:2187. [PMID: 33846346 PMCID: PMC8041803 DOI: 10.1038/s41467-021-22484-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/16/2021] [Indexed: 02/08/2023] Open
Abstract
The RNA-sensing pathway contributes to type I interferon (IFN) production induced by DNA damaging agents. However, the potential involvement of RNA sensors in DNA repair is unknown. Here, we found that retinoic acid-inducible gene I (RIG-I), a key cytosolic RNA sensor that recognizes RNA virus and initiates the MAVS-IRF3-type I IFN signaling cascade, is recruited to double-stranded breaks (DSBs) and suppresses non-homologous end joining (NHEJ). Mechanistically, RIG-I interacts with XRCC4, and the RIG-I/XRCC4 interaction impedes the formation of XRCC4/LIG4/XLF complex at DSBs. High expression of RIG-I compromises DNA repair and sensitizes cancer cells to irradiation treatment. In contrast, depletion of RIG-I renders cells resistant to irradiation in vitro and in vivo. In addition, this mechanism suggests a protective role of RIG-I in hindering retrovirus integration into the host genome by suppressing the NHEJ pathway. Reciprocally, XRCC4, while suppressed for its DNA repair function, has a critical role in RIG-I immune signaling through RIG-I interaction. XRCC4 promotes RIG-I signaling by enhancing oligomerization and ubiquitination of RIG-I, thereby suppressing RNA virus replication in host cells. In vivo, silencing XRCC4 in mouse lung promotes influenza virus replication in mice and these mice display faster body weight loss, poorer survival, and a greater degree of lung injury caused by influenza virus infection. This reciprocal regulation of RIG-I and XRCC4 reveals a new function of RIG-I in suppressing DNA repair and virus integration into the host genome, and meanwhile endues XRCC4 with a crucial role in potentiating innate immune response, thereby helping host to prevail in the battle against virus.
Collapse
Affiliation(s)
- Guijie Guo
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Ming Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Xiaochen Gao
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Bibo Zhu
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jinzhou Huang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Xinyi Tu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Wootae Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Fei Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Qin Zhou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Shouhai Zhu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Zheming Wu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Yuanliang Yan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Yong Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Xiangyu Zeng
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Qian Zhu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Ping Yin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Kuntian Luo
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jie Sun
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Min Deng
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| | - Zhenkun Lou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Elliott JL, Kutluay SB. Going beyond Integration: The Emerging Role of HIV-1 Integrase in Virion Morphogenesis. Viruses 2020; 12:E1005. [PMID: 32916894 PMCID: PMC7551943 DOI: 10.3390/v12091005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022] Open
Abstract
The HIV-1 integrase enzyme (IN) plays a critical role in the viral life cycle by integrating the reverse-transcribed viral DNA into the host chromosome. This function of IN has been well studied, and the knowledge gained has informed the design of small molecule inhibitors that now form key components of antiretroviral therapy regimens. Recent discoveries unveiled that IN has an under-studied yet equally vital second function in human immunodeficiency virus type 1 (HIV-1) replication. This involves IN binding to the viral RNA genome in virions, which is necessary for proper virion maturation and morphogenesis. Inhibition of IN binding to the viral RNA genome results in mislocalization of the viral genome inside the virus particle, and its premature exposure and degradation in target cells. The roles of IN in integration and virion morphogenesis share a number of common elements, including interaction with viral nucleic acids and assembly of higher-order IN multimers. Herein we describe these two functions of IN within the context of the HIV-1 life cycle, how IN binding to the viral genome is coordinated by the major structural protein, Gag, and discuss the value of targeting the second role of IN in virion morphogenesis.
Collapse
Affiliation(s)
| | - Sebla B. Kutluay
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA;
| |
Collapse
|
16
|
Anisenko A, Kan M, Shadrina O, Brattseva A, Gottikh M. Phosphorylation Targets of DNA-PK and Their Role in HIV-1 Replication. Cells 2020; 9:E1907. [PMID: 32824372 PMCID: PMC7464883 DOI: 10.3390/cells9081907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The DNA dependent protein kinase (DNA-PK) is a trimeric nuclear complex consisting of a large protein kinase and the Ku heterodimer. The kinase activity of DNA-PK is required for efficient repair of DNA double-strand breaks (DSB) by non-homologous end joining (NHEJ). We also showed that the kinase activity of DNA-PK is essential for post-integrational DNA repair in the case of HIV-1 infection. Besides, DNA-PK is known to participate in such cellular processes as protection of mammalian telomeres, transcription, and some others where the need for its phosphorylating activity is not clearly elucidated. We carried out a systematic search and analysis of DNA-PK targets described in the literature and identified 67 unique DNA-PK targets phosphorylated in response to various in vitro and/or in vivo stimuli. A functional enrichment analysis of DNA-PK targets and determination of protein-protein associations among them were performed. For 27 proteins from these 67 DNA-PK targets, their participation in the HIV-1 life cycle was demonstrated. This information may be useful for studying the functioning of DNA-PK in various cellular processes, as well as in various stages of HIV-1 replication.
Collapse
Affiliation(s)
- Andrey Anisenko
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Kan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Olga Shadrina
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Anna Brattseva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Gottikh
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
| |
Collapse
|
17
|
Knyazhanskaya E, Anisenko A, Shadrina O, Kalinina A, Zatsepin T, Zalevsky A, Mazurov D, Gottikh M. NHEJ pathway is involved in post-integrational DNA repair due to Ku70 binding to HIV-1 integrase. Retrovirology 2019; 16:30. [PMID: 31690330 PMCID: PMC6833283 DOI: 10.1186/s12977-019-0492-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Background HIV-1 integration results in genomic DNA gaps that are repaired by cellular DNA repair pathways. This step of the lentiviral life cycle remains poorly understood despite its crucial importance for successful replication. We and others reported that Ku70 protein of the non-homologous end joining pathway (NHEJ) directly binds HIV-1 integrase (IN). Here, we studied the importance of this interaction for post-integrational gap repair and the recruitment of NHEJ factors in this process. Results We engineered HIV-based pseudovirus with mutant IN defective in Ku70 binding and generated heterozygous Ku70, Ku80 and DNA-PKcs human knockout (KO) cells using CRISPR/Cas9. KO of either of these proteins or inhibition of DNA-PKcs catalytic activity substantially decreased the infectivity of HIV-1 with native IN but not with the mutant one. We used a recently developed qPCR assay for the measurement of gap repair efficiency to show that HIV-1 with mutant IN was defective in DNA post-integrational repair, whereas the wild type virus displayed such a defect only when NHEJ system was disrupted in any way. This effect was present in CRISPR/Cas9 modified 293T cells, in Jurkat and CEM lymphoid lines and in primary human PBMCs. Conclusions Our data provide evidence that IN recruits DNA-PK to the site of HIV-1 post-integrational repair due to Ku70 binding—a novel finding that explains the involvement of DNA-PK despite the absence of free double stranded DNA breaks. In addition, our data clearly indicate the importance of interactions between HIV-1 IN and Ku70 in HIV-1 replication at the post-integrational repair step.
Collapse
Affiliation(s)
- Ekaterina Knyazhanskaya
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia. .,Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Andrey Anisenko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Olga Shadrina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Anastasia Kalinina
- Federal State Budgetary Institution « N.N. Blokhin National Medical Research Center of Oncology » of the Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Timofei Zatsepin
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia.,Skolkovo Institute of Science and Technology, Skolkovo, 121205, Russia
| | - Arthur Zalevsky
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Dmitriy Mazurov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, RAS, Moscow, 119334, Russia.,NRC Institute of Immunology FMBA of Russia, Moscow, 115478, Russia
| | - Marina Gottikh
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
18
|
Tanaka M, Tanaka K, Ida C, Oue A, Yamashita S, Yao J, Takenouchi N, Miwa M. Conflicting effects of poly(ADP-ribose) polymerase inhibitor on cell-mediated and virion-mediated HTLV-1 infection. Virus Res 2019; 270:197653. [PMID: 31299194 DOI: 10.1016/j.virusres.2019.197653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/16/2019] [Accepted: 07/08/2019] [Indexed: 11/18/2022]
Abstract
Adult T-cell leukemia and human T-cell leukemia virus type 1 (HTLV-1) - associated myelopathy/tropical spastic paraparesis, which develop after HTLV-1 infection, are difficult to cure. In particular, the mode of HTLV-1 propagation is not well understood. Poly (ADP-ribose) polymerase-1 is reported to be a co-activator of HTLV-1 Tax protein; however, the effects of polyADP-ribosylation on infectivity of HTLV-1 have not been fully clarified. We studied the effects of a PARP inhibitor on two modes of HTLV-1 transmission: through cell adhesion between MT-2 cells (an HTLV-1-infected cell line) and uninfected cells and through virus particles produced by HTLV-1-producing c77 cells. Although the PARP inhibitor decreased HTLV-1 infection through cell adhesion, it increased HTLV-1 infection through virion production and caused apoptosis of HTLV-1-infected cells. Thus, careful consideration is required for clinical application of PARP inhibitors in HTLV-1 patients.
Collapse
Affiliation(s)
- Masakazu Tanaka
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan; Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Kagoshima 890-8544, Japan.
| | - Koji Tanaka
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan; Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Chieri Ida
- Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan; College of Nagoya Women's University, Nagoya, Aichi, 467-8610, Japan
| | - Atsushi Oue
- Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Sachiko Yamashita
- Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Jinchun Yao
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Norihiro Takenouchi
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Masanao Miwa
- Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan; Institute of Basic Medical Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
19
|
|
20
|
Majer C, Schüssler JM, König R. Intertwined: SAMHD1 cellular functions, restriction, and viral evasion strategies. Med Microbiol Immunol 2019; 208:513-529. [PMID: 30879196 DOI: 10.1007/s00430-019-00593-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/08/2019] [Indexed: 01/01/2023]
Abstract
SAMHD1 was initially described for its ability to efficiently restrict HIV-1 replication in myeloid cells and resting CD4+ T cells. However, a growing body of evidence suggests that SAMHD1-mediated restriction is by far not limited to lentiviruses, but seems to be a general concept that applies to most retroviruses and at least a number of DNA viruses. SAMHD1 anti-viral activity was long believed to be solely due to its ability to deplete cellular dNTPs by enzymatic degradation. However, since its discovery, several new functions have been attributed to SAMHD1. It has been demonstrated to bind nucleic acids, to modulate innate immunity, as well as to participate in the DNA damage response and resolution of stalled replication forks. Consequently, it is likely that SAMHD1-mediated anti-viral activity is not or not exclusively mediated through its dNTPase activity. Therefore, in this review, we summarize current knowledge on SAMHD1 cellular functions and systematically discuss how these functions could contribute to the restriction of a broad range of viruses besides retroviruses: herpesviruses, poxviruses and hepatitis B virus. Furthermore, we aim to highlight different ways how viruses counteract SAMHD1-mediated restriction to bypass the SAMHD1-mediated block to viral infection.
Collapse
Affiliation(s)
- Catharina Majer
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany
| | | | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany. .,Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA. .,German Center for Infection Research (DZIF), 63225, Langen, Germany. .,Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany.
| |
Collapse
|
21
|
A qPCR assay for measuring the post-integrational DNA repair in HIV-1 replication. J Virol Methods 2018; 262:12-19. [PMID: 30219707 DOI: 10.1016/j.jviromet.2018.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 01/13/2023]
Abstract
The post-integrational gap repair is a critical and poorly studied stage of the lentiviral life cycle. It might be performed by various cellular DNA repair pathways but the exact mechanism of the repair process has not yet been described. One of the reasons for that is the lack of a functional quantitative assay that could precisely measure the amount of integrated viral DNA that has completed the post-integrational gap repair stage. Here, we present an approach that is based on a widely used Alu-specific PCR for the estimation of integrated viral DNA but includes several steps that allow discrimination between integrated-repaired and integrated-unrepaired viral DNA forms. We used the approach for the estimation of the kinetics of gap repair in a viral vector system and showed that the gap repair process starts at 17 h post infection and lasts 10 more hours. We also showed that the addition of Nu7441 - a small molecule inhibitor of DNA-breaks sensor kinase in the non-homologous end joining DNA repair pathway - specifically inhibits the gap repair process while having no influence on the integration itself.
Collapse
|
22
|
Border collies of the genome: domestication of an autonomous retrovirus-like transposon. Curr Genet 2018; 65:71-78. [PMID: 29931377 DOI: 10.1007/s00294-018-0857-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
Abstract
Retrotransposons often spread rapidly through eukaryotic genomes until they are neutralized by host-mediated silencing mechanisms, reduced by recombination and mutation, and lost or transformed into benevolent entities. But the Ty1 retrotransposon appears to have been domesticated to guard the genome of Saccharomyces cerevisiae.
Collapse
|
23
|
Kuo HH, Ahmad R, Lee GQ, Gao C, Chen HR, Ouyang Z, Szucs MJ, Kim D, Tsibris A, Chun TW, Battivelli E, Verdin E, Rosenberg ES, Carr SA, Yu XG, Lichterfeld M. Anti-apoptotic Protein BIRC5 Maintains Survival of HIV-1-Infected CD4 + T Cells. Immunity 2018; 48:1183-1194.e5. [PMID: 29802019 DOI: 10.1016/j.immuni.2018.04.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/17/2018] [Accepted: 04/02/2018] [Indexed: 01/16/2023]
Abstract
HIV-1 infection of CD4+ T cells leads to cytopathic effects and cell demise, which is counter to the observation that certain HIV-1-infected cells possess a remarkable long-term stability and can persist lifelong in infected individuals treated with suppressive antiretroviral therapy (ART). Using quantitative mass spectrometry-based proteomics, we showed that HIV-1 infection activated cellular survival programs that were governed by BIRC5, a molecular inhibitor of cell apoptosis that is frequently overexpressed in malignant cells. BIRC5 and its upstream regulator OX40 were upregulated in productively and latently infected CD4+ T cells and were functionally involved in maintaining their viability. Moreover, OX40-expressing CD4+ T cells from ART-treated patients were enriched for clonally expanded HIV-1 sequences, and pharmacological inhibition of BIRC5 resulted in a selective decrease of HIV-1-infected cells in vitro. Together, these findings suggest that BIRC5 supports long-term survival of HIV-1-infected cells and may lead to clinical strategies to reduce persisting viral reservoirs.
Collapse
Affiliation(s)
- Hsiao-Hsuan Kuo
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Guinevere Q Lee
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Hsiao-Rong Chen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zhengyu Ouyang
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew J Szucs
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Dhohyung Kim
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Athe Tsibris
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tae-Wook Chun
- National Institute of Allergies and Infectious Diseases, Bethesda, MD 20892, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Eric S Rosenberg
- Infectious Disease Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
24
|
Hou J, Xu T, Su D, Wu Y, Cheng L, Wang J, Zhou Z, Wang Y. RNA-Seq Reveals Extensive Transcriptional Response to Heat Stress in the Stony Coral Galaxea fascicularis. Front Genet 2018; 9:37. [PMID: 29487614 PMCID: PMC5816741 DOI: 10.3389/fgene.2018.00037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/29/2018] [Indexed: 11/13/2022] Open
Abstract
Galaxea fascicularis, a stony coral belonging to family Oculinidae, is widely distributed in Red Sea, the Gulf of Aden and large areas of the Indo-Pacific oceans. So far there is a lack of gene expression knowledge concerning this massive coral. In the present study, G. fascicularis was subjected to heat stress at 32.0 ± 0.5°C in the lab, we found that the density of symbiotic zooxanthellae decreased significantly; meanwhile apparent bleaching and tissue lysing were observed at 10 h and 18 h after heat stress. The transcriptome responses were investigated in the stony coral G. fascicularis during heat bleaching using RNA-seq. A total of 42,028 coral genes were assembled from over 439 million reads. Gene expressions were compared at 10 and 18 h after heat stress. The significantly upregulated genes found in the Control_10h vs. Heat_10h comparison, presented mainly in GO terms related with DNA integration and unfolded protein response; and for the Control_18h vs. Heat_18h comparison, the GO terms include DNA integration. In addition, comparison between groups of Control_10h vs. Heat_10h and Control_18h vs. Heat_18h revealed that 125 genes were significantly upregulated in common between the two groups, whereas 21 genes were significantly downregulated in common, all these differentially expressed genes were found to be involved in stress response, DNA integration and unfolded protein response. Taken together, our results suggest that high temperature could activate the stress response at the early stage, and subsequently induce the bleaching and lysing through DNA integration and unfolded protein response, which are able to disrupt the balance of coral-zooxanthella symbiosis in the stony coral G. fascicularis.
Collapse
Affiliation(s)
- Jing Hou
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Tao Xu
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Dingjia Su
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Ying Wu
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Li Cheng
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Jun Wang
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Zhi Zhou
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| | - Yan Wang
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Key Laboratory of Tropical Biological Resources of Ministry of Education, Ocean College, Hainan University, Haikou, China
| |
Collapse
|
25
|
Mlcochova P, Caswell SJ, Taylor IA, Towers GJ, Gupta RK. DNA damage induced by topoisomerase inhibitors activates SAMHD1 and blocks HIV-1 infection of macrophages. EMBO J 2018; 37:50-62. [PMID: 29084722 PMCID: PMC5753034 DOI: 10.15252/embj.201796880] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 12/15/2022] Open
Abstract
We report that DNA damage induced by topoisomerase inhibitors, including etoposide (ETO), results in a potent block to HIV-1 infection in human monocyte-derived macrophages (MDM). SAMHD1 suppresses viral reverse transcription (RT) through depletion of cellular dNTPs but is naturally switched off by phosphorylation in a subpopulation of MDM found in a G1-like state. We report that SAMHD1 was activated by dephosphorylation following ETO treatment, along with loss of expression of MCM2 and CDK1, and reduction in dNTP levels. Suppression of infection occurred after completion of viral DNA synthesis, at the step of 2LTR circle and provirus formation. The ETO-induced block was completely rescued by depletion of SAMHD1 in MDM Concordantly, infection by HIV-2 and SIVsm encoding the SAMHD1 antagonist Vpx was insensitive to ETO treatment. The mechanism of DNA damage-induced blockade of HIV-1 infection involved activation of p53, p21, decrease in CDK1 expression, and SAMHD1 dephosphorylation. Therefore, topoisomerase inhibitors regulate SAMHD1 and HIV permissivity at a post-RT step, revealing a mechanism by which the HIV-1 reservoir may be limited by chemotherapeutic drugs.
Collapse
Affiliation(s)
| | - Sarah J Caswell
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, UK
| | - Ian A Taylor
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, UK
| | | | - Ravindra K Gupta
- Division of Infection and Immunity, UCL, London, UK
- Africa Health Research Institute, Durban, KwaZulu Natal, South Africa
| |
Collapse
|
26
|
Smith EA, Krumpelbeck EF, Jegga AG, Greis KD, Ali AM, Meetei AR, Wells SI. The nuclear DEK interactome supports multi-functionality. Proteins 2018; 86:88-97. [PMID: 29082557 PMCID: PMC5730476 DOI: 10.1002/prot.25411] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/20/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023]
Abstract
DEK is an oncoprotein that is overexpressed in many forms of cancer and participates in numerous cellular pathways. Of these different pathways, relevant interacting partners and functions of DEK are well described in regard to the regulation of chromatin structure, epigenetic marks, and transcription. Most of this understanding was derived by investigating DNA-binding and chromatin processing capabilities of the oncoprotein. To facilitate the generation of mechanism-driven hypotheses regarding DEK activities in underexplored areas, we have developed the first DEK interactome model using tandem-affinity purification and mass spectrometry. With this approach, we identify IMPDH2, DDX21, and RPL7a as novel DEK binding partners, hinting at new roles for the oncogene in de novo nucleotide biosynthesis and ribosome formation. Additionally, a hydroxyurea-specific interaction with replication protein A (RPA) was observed, suggesting that a DEK-RPA complex may form in response to DNA replication fork stalling. Taken together, these findings highlight diverse activities for DEK across cellular pathways and support a model wherein this molecule performs a plethora of functions.
Collapse
Affiliation(s)
- Eric A. Smith
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Eric F. Krumpelbeck
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45219, USA
| | - Kenneth D. Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine; Cincinnati, OH 45219, USA
| | - Abdullah M. Ali
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Amom R. Meetei
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Susanne I. Wells
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| |
Collapse
|
27
|
Abstract
My laboratory investigations have been driven by an abiding interest in understanding the consequences of genetic rearrangement in evolution and disease, and in using viruses to elucidate fundamental mechanisms in biology. Starting with bacteriophages and moving to the retroviruses, my use of the tools of genetics, molecular biology, biochemistry, and biophysics has spanned more than half a century-from the time when DNA structure was just discovered to the present day of big data and epigenetics. Both riding and contributing to the successive waves of technology, my laboratory has elucidated fundamental mechanisms in DNA replication, repair, and recombination. We have made substantial contributions in the area of retroviral oncogenesis, delineated mechanisms that control retroviral gene expression, and elucidated critical details of the structure and function of the retroviral enzymes-reverse transcriptase, protease, and integrase-and have had the satisfaction of knowing that the fundamental knowledge gained from these studies contributed important groundwork for the eventual development of antiviral drugs to treat AIDS. While pursuing laboratory research as a principal investigator, I have also been a science administrator-moving from laboratory head to department chair and, finally, to institute director. In addition, I have undertaken a number of community service, science-related "extracurricular" activities during this time. Filling all of these roles, while being a wife and mother, has required family love and support, creative management, and, above all, personal flexibility-with not too much long-term planning. I hope that this description of my journey, with various roles, obstacles, and successes, will be both interesting and informative, especially to young female scientists.
Collapse
Affiliation(s)
- Anna Marie Ann Skalka
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111;
| |
Collapse
|
28
|
Potent Inhibition of HIV-1 Replication in Resting CD4 T Cells by Resveratrol and Pterostilbene. Antimicrob Agents Chemother 2017; 61:AAC.00408-17. [PMID: 28652233 DOI: 10.1128/aac.00408-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
HIV-1 infection of resting CD4 T cells plays a crucial and numerically dominant role during virus transmission at mucosal sites and during subsequent acute replication and T cell depletion. Resveratrol and pterostilbene are plant stilbenoids associated with several health-promoting benefits. Resveratrol has been shown to inhibit the replication of several viruses, including herpes simplex viruses 1 and 2, papillomaviruses, severe acute respiratory syndrome virus, and influenza virus. Alone, resveratrol does not inhibit HIV-1 infection of activated T cells, but it does synergize with nucleoside reverse transcriptase inhibitors in these cells to inhibit reverse transcription. Here, we demonstrate that resveratrol and pterostilbene completely block HIV-1 infection at a low micromolar dose in resting CD4 T cells, primarily at the reverse transcription step. The anti-HIV effect was fully reversed by exogenous deoxynucleosides and Vpx, an HIV-1 and simian immunodeficiency virus protein that increases deoxynucleoside triphosphate (dNTP) levels. These findings are consistent with the reported ability of resveratrol to inhibit ribonucleotide reductase and to lower dNTP levels in cells. This study supports the potential use of resveratrol, pterostilbene, or related compounds as adjuvants in anti-HIV preexposure prophylaxis (PrEP) formulations.
Collapse
|
29
|
Characterization of HIV-1 integrase interaction with human Ku70 protein and initial implications for drug targeting. Sci Rep 2017; 7:5649. [PMID: 28717247 PMCID: PMC5514147 DOI: 10.1038/s41598-017-05659-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/01/2017] [Indexed: 11/27/2022] Open
Abstract
Human Ku70/Ku80 protein is known to influence HIV-1 replication. One of the possible reasons may be the protection of integrase from proteasomal degradation by Ku70 subunit. We demonstrated that recombinant HIV-1 integrase and Ku70 form a stable complex, while no interaction of Ku70 with integrase from prototype foamy virus was observed. By analyzing protein subdomains we determined two binding sites in the structure of both Ku70 and integrase: the 51–160 a.a. region of integrase interacts with residues 251–438 of Ku70, whereas Ku70 N-terminal domain (1–250 a.a.) contacts an α6-helix in the 200–220 a.a. integrase region. Single substitutions within integrase (E212A or L213A) block the interaction with Ku70 thus indicating that the binding site formed by the 200–220 a.a. integrase region is crucial for complex formation. E212A/L213A substitutions decreased the integrase capacity to bind Ku70 in HEK293T cells. A conjugate of 2′-ОMe-GGUUUUUGUGU oligonucleotide with eosin is shown by molecular modeling to shield integrase residues E212/L213 and is effective in blocking complex formation of Ku70 with integrase what makes the complex between α6-helix and Ku70(1–250) a possible target for drug development.
Collapse
|
30
|
Transcriptomic Analysis Implicates the p53 Signaling Pathway in the Establishment of HIV-1 Latency in Central Memory CD4 T Cells in an In Vitro Model. PLoS Pathog 2016; 12:e1006026. [PMID: 27898737 PMCID: PMC5127598 DOI: 10.1371/journal.ppat.1006026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/26/2016] [Indexed: 12/19/2022] Open
Abstract
The search for an HIV-1 cure has been greatly hindered by the presence of a viral reservoir that persists despite antiretroviral therapy (ART). Studies of HIV-1 latency in vivo are also complicated by the low proportion of latently infected cells in HIV-1 infected individuals. A number of models of HIV-1 latency have been developed to examine the signaling pathways and viral determinants of latency and reactivation. A primary cell model of HIV-1 latency, which incorporates the generation of primary central memory CD4 T cells (TCM), full-length virus infection (HIVNL4-3) and ART to suppress virus replication, was used to investigate the establishment of HIV latency using RNA-Seq. Initially, an investigation of host and viral gene expression in the resting and activated states of this model indicated that the resting condition was reflective of a latent state. Then, a comparison of the host transcriptome between the uninfected and latently infected conditions of this model identified 826 differentially expressed genes, many of which were related to p53 signaling. Inhibition of the transcriptional activity of p53 by pifithrin-α during HIV-1 infection reduced the ability of HIV-1 to be reactivated from its latent state by an unknown mechanism. In conclusion, this model may be used to screen latency reversing agents utilized in shock and kill approaches to cure HIV, to search for cellular markers of latency, and to understand the mechanisms by which HIV-1 establishes latency.
Collapse
|
31
|
Knyazhanskaya ES, Shadrina OA, Anisenko AN, Gottikh MB. Role of DNA-dependent protein kinase in the HIV-1 replication cycle. Mol Biol 2016. [DOI: 10.1134/s0026893316040075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
32
|
Abstract
The integration of a DNA copy of the viral RNA genome into host chromatin is the defining step of retroviral replication. This enzymatic process is catalyzed by the virus-encoded integrase protein, which is conserved among retroviruses and LTR-retrotransposons. Retroviral integration proceeds via two integrase activities: 3'-processing of the viral DNA ends, followed by the strand transfer of the processed ends into host cell chromosomal DNA. Herein we review the molecular mechanism of retroviral DNA integration, with an emphasis on reaction chemistries and architectures of the nucleoprotein complexes involved. We additionally discuss the latest advances on anti-integrase drug development for the treatment of AIDS and the utility of integrating retroviral vectors in gene therapy applications.
Collapse
Affiliation(s)
- Paul Lesbats
- Clare Hall Laboratories, The Francis Crick Institute , Blanche Lane, South Mimms, EN6 3LD, U.K
| | - Alan N Engelman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School , 450 Brookline Avenue, Boston, Massachusetts 02215 United States
| | - Peter Cherepanov
- Clare Hall Laboratories, The Francis Crick Institute , Blanche Lane, South Mimms, EN6 3LD, U.K.,Imperial College London , St-Mary's Campus, Norfolk Place, London, W2 1PG, U.K
| |
Collapse
|
33
|
Integration of HIV in the Human Genome: Which Sites Are Preferential? A Genetic and Statistical Assessment. Int J Genomics 2016; 2016:2168590. [PMID: 27294106 PMCID: PMC4880676 DOI: 10.1155/2016/2168590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/24/2016] [Indexed: 12/17/2022] Open
Abstract
Chromosomal fragile sites (FSs) are loci where gaps and breaks may occur and are preferential integration targets for some viruses, for example, Hepatitis B, Epstein-Barr virus, HPV16, HPV18, and MLV vectors. However, the integration of the human immunodeficiency virus (HIV) in Giemsa bands and in FSs is not yet completely clear. This study aimed to assess the integration preferences of HIV in FSs and in Giemsa bands using an in silico study. HIV integration positions from Jurkat cells were used and two nonparametric tests were applied to compare HIV integration in dark versus light bands and in FS versus non-FS (NFSs). The results show that light bands are preferential targets for integration of HIV-1 in Jurkat cells and also that it integrates with equal intensity in FSs and in NFSs. The data indicates that HIV displays different preferences for FSs compared to other viruses. The aim was to develop and apply an approach to predict the conditions and constraints of HIV insertion in the human genome which seems to adequately complement empirical data.
Collapse
|
34
|
Activation of the DNA Damage Response by RNA Viruses. Biomolecules 2016; 6:2. [PMID: 26751489 PMCID: PMC4808796 DOI: 10.3390/biom6010002] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
RNA viruses are a genetically diverse group of pathogens that are responsible for some of the most prevalent and lethal human diseases. Numerous viruses introduce DNA damage and genetic instability in host cells during their lifecycles and some species also manipulate components of the DNA damage response (DDR), a complex and sophisticated series of cellular pathways that have evolved to detect and repair DNA lesions. Activation and manipulation of the DDR by DNA viruses has been extensively studied. It is apparent, however, that many RNA viruses can also induce significant DNA damage, even in cases where viral replication takes place exclusively in the cytoplasm. DNA damage can contribute to the pathogenesis of RNA viruses through the triggering of apoptosis, stimulation of inflammatory immune responses and the introduction of deleterious mutations that can increase the risk of tumorigenesis. In addition, activation of DDR pathways can contribute positively to replication of viral RNA genomes. Elucidation of the interactions between RNA viruses and the DDR has provided important insights into modulation of host cell functions by these pathogens. This review summarises the current literature regarding activation and manipulation of the DDR by several medically important RNA viruses.
Collapse
|
35
|
HERD OLIVIA, FRANCIES FLAVIA, KOTZEN JEFFREY, SMITH TRUDY, NXUMALO ZWIDE, MULLER XANTHENE, SLABBERT JACOBUS, VRAL ANNE, BAEYENS ANS. Chromosomal radiosensitivity of human immunodeficiency virus positive/negative cervical cancer patients in South Africa. Mol Med Rep 2016; 13:130-6. [PMID: 26549042 PMCID: PMC4686097 DOI: 10.3892/mmr.2015.4504] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/28/2015] [Indexed: 12/01/2022] Open
Abstract
Cervical cancer is the second most common cancer amongst South African women and is the leading cause of cancer-associated mortality in this region. Several international studies on radiation‑induced DNA damage in lymphocytes of cervical cancer patients have remained inconclusive. Despite the high incidence of cervical cancer in South Africa, and the extensive use of radiotherapy to treat it, the chromosomal radiosensitivity of South African cervical cancer patients has not been studied to date. Since a high number of these patients are human immunodeficiency virus (HIV)‑positive, the effect of HIV infection on chromosomal radiosensitivity was also investigated. Blood samples from 35 cervical cancer patients (20 HIV‑negative and 15 HIV‑positive) and 20 healthy controls were exposed to X‑rays at doses of 6 MV of 2 and 4 Gy in vitro. Chromosomal radiosensitivity was assessed using the micronucleus (MN) assay. MN scores were obtained using the Metafer 4 platform, an automated microscopic system. Three scoring methods of the MNScore module of Metafer were applied and compared. Cervical cancer patients had higher MN values than healthy controls, with HIV‑positive patients having the highest MN values. Differences between groups were significant when using a scoring method that corrects for false positive and false negative MN. The present study suggested increased chromosomal radiosensitivity in HIV-positive South African cervical cancer patients.
Collapse
Affiliation(s)
- OLIVIA HERD
- Department of Radiation Biophysics, NRF-iThemba LABS, Somerset West 7129, South Africa
- Department of Radiation Sciences, University of Witwatersrand, Johannesburg 2193, South Africa
| | - FLAVIA FRANCIES
- Department of Radiation Biophysics, NRF-iThemba LABS, Somerset West 7129, South Africa
- Department of Radiation Sciences, University of Witwatersrand, Johannesburg 2193, South Africa
| | - JEFFREY KOTZEN
- Department of Radiation Oncology, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg 2193, South Africa
| | - TRUDY SMITH
- Department of Obstetrics and Gynaecology, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg 2193, South Africa
| | - ZWIDE NXUMALO
- Department of Obstetrics and Gynaecology, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg 2193, South Africa
| | - XANTHENE MULLER
- Department of Radiation Biophysics, NRF-iThemba LABS, Somerset West 7129, South Africa
| | - JACOBUS SLABBERT
- Department of Radiation Biophysics, NRF-iThemba LABS, Somerset West 7129, South Africa
| | - ANNE VRAL
- Department of Basic Medical Sciences, Ghent University, Ghent B-9000, Belgium
| | - ANS BAEYENS
- Department of Radiation Biophysics, NRF-iThemba LABS, Somerset West 7129, South Africa
- Department of Radiation Sciences, University of Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
36
|
Abstract
The retroviral integrases are virally encoded, specialized recombinases that catalyze the insertion of viral DNA into the host cell's DNA, a process that is essential for virus propagation. We have learned a great deal since the existence of an integrated form of retroviral DNA (the provirus) was first proposed by Howard Temin in 1964. Initial studies focused on the genetics and biochemistry of avian and murine virus DNA integration, but the pace of discovery increased substantially with advances in technology, and an influx of investigators focused on the human immunodeficiency virus. We begin with a brief account of the scientific landscape in which some of the earliest discoveries were made, and summarize research that led to our current understanding of the biochemistry of integration. A more detailed account of recent analyses of integrase structure follows, as they have provided valuable insights into enzyme function and raised important new questions.
Collapse
Affiliation(s)
- Mark D Andrake
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111; ,
| | - Anna Marie Skalka
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111; ,
| |
Collapse
|
37
|
Qi S, Fang Z, Wang D, Menendez P, Yao K, Ji J. Concise Review: Induced Pluripotency by Defined Factors: Prey of Oxidative Stress. Stem Cells 2015; 33:1371-6. [DOI: 10.1002/stem.1946] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/13/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Suxia Qi
- Center of Stem Cell and Regenerative Medicine; School of Medicine; Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
| | - Zhi Fang
- Center of Stem Cell and Regenerative Medicine; School of Medicine; Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
- Eye Institute of Zhejiang University; Eye Center of Second Affiliated Hospital of Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
| | - Danli Wang
- Center of Stem Cell and Regenerative Medicine; School of Medicine; Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute and Cell Therapy Program of School of Medicine; University of Barcelona; Barcelona Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA); Barcelona Spain
| | - Ke Yao
- Eye Institute of Zhejiang University; Eye Center of Second Affiliated Hospital of Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
| | - Junfeng Ji
- Center of Stem Cell and Regenerative Medicine; School of Medicine; Zhejiang University; Hangzhou Zhejiang Province People's Republic of China
- Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine; Hangzhou People's Republic of China
| |
Collapse
|
38
|
Abstract
Retroviruses and LTR retrotransposons are transposable elements that encapsidate the RNAs that are intermediates in the transposition of DNA copies of their genomes (proviruses), from one cell (or one locus) to another. Mechanistic similarities in DNA transposase enzymes and retroviral/retrotransposon integrases underscore the close evolutionary relationship among these elements. The retroviruses are very ancient infectious agents, presumed to have evolved from Ty3/Gypsy LTR retrotransposons (1), and DNA copies of their sequences can be found embedded in the genomes of most, if not all, members of the tree of life. All retroviruses share a specific gene arrangement and similar replication strategies. However, given their ancestries and occupation of diverse evolutionary niches, it should not be surprising that unique sequences have been acquired in some retroviral genomes and that the details of the mechanism by which their transposition is accomplished can vary. While every step in the retrovirus lifecycle is, in some sense, relevant to transposition, this Chapter focuses mainly on the early phase of retroviral replication, during which viral DNA is synthesized and integrated into its host genome. Some of the initial studies that set the stage for current understanding are highlighted, as well as more recent findings obtained through use of an ever-expanding technological toolbox including genomics, proteomics, and siRNA screening. Persistence in the area of structural biology has provided new insight into conserved mechanisms as well as variations in detail among retroviruses, which can also be instructive.
Collapse
Affiliation(s)
- Anna Marie Skalka
- Fox Chase Cancer Center 333 Cottman Avenue Philadelphia, PA 19111 United States 2157282192 2157282778 (fax)
| |
Collapse
|
39
|
Fontes FL, Pinheiro DML, Oliveira AHSD, Oliveira RKDM, Lajus TBP, Agnez-Lima LF. Role of DNA repair in host immune response and inflammation. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 763:246-57. [PMID: 25795123 DOI: 10.1016/j.mrrev.2014.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/28/2022]
Abstract
In recent years, the understanding of how DNA repair contributes to the development of innate and acquired immunity has emerged. The DNA damage incurred during the inflammatory response triggers the activation of DNA repair pathways, which are required for host-cell survival. Here, we reviewed current understanding of the mechanism by which DNA repair contributes to protection against the oxidized DNA damage generated during infectious and inflammatory diseases and its involvement in innate and adaptive immunity. We discussed the functional role of DNA repair enzymes in the immune activation and the relevance of these processes to: transcriptional regulation of cytokines and other genes involved in the inflammatory response; V(D)J recombination; class-switch recombination (CSR); and somatic hypermutation (SHM). These three last processes of DNA damage repair are required for effective humoral adaptive immunity, creating genetic diversity in developing T and B cells. Furthermore, viral replication is also dependent on host DNA repair mechanisms. Therefore, the elucidation of the pathways of DNA damage and its repair that activate innate and adaptive immunity will be important for a better understanding of the immune and inflammatory disorders and developing new therapeutic interventions for treatment of these diseases and for improving their outcome.
Collapse
Affiliation(s)
- Fabrícia Lima Fontes
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brazil.
| | - Daniele Maria Lopes Pinheiro
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brazil.
| | - Ana Helena Sales de Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brazil.
| | | | - Tirzah Braz Petta Lajus
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brazil; Liga Contra o Cancer, Natal, RN, Brazil.
| | | |
Collapse
|
40
|
Wang DJ, Ratnam NM, Byrd JC, Guttridge DC. NF-κB functions in tumor initiation by suppressing the surveillance of both innate and adaptive immune cells. Cell Rep 2014; 9:90-103. [PMID: 25263557 PMCID: PMC4882153 DOI: 10.1016/j.celrep.2014.08.049] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 06/25/2014] [Accepted: 08/21/2014] [Indexed: 12/11/2022] Open
Abstract
NF-κB is considered a major contributor to tumor development, but how this factor functions in the initial stages of oncogenesis is not clear. In a model of Ras-induced transformation, we probed NF-κB function as preneoplastic cells formed tumors in mice. As previously shown, the p65 subunit of NF-κB acts as a tumor suppressor in normal cells by sustaining senescence following DNA damage. Our current data reveal that, following immortalization, p65 switches to an oncogene by counteracting the surveillance properties of immune cells. NF-κB exerts this effect by protecting transformed cells against macrophage-derived proapoptotic factors, tumor necrosis factor, and nitric oxide. Additionally, NF-κB acts through transforming growth factor beta (TGF-β) to mitigate T cell cytotoxicity and other factors to expand myeloid-derived suppressor cells. Together, these data suggest that NF-κB functions in the early stages of transformation by suppressing immune surveillance of both innate and adaptive immune cells, information that may be useful for targeted immunotherapies.
Collapse
Affiliation(s)
- David J Wang
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA; Department of Internal Medicine, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nivedita M Ratnam
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - John C Byrd
- Department of Internal Medicine, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Internal Medicine, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
41
|
Chatzinikolaou G, Karakasilioti I, Garinis GA. DNA damage and innate immunity: links and trade-offs. Trends Immunol 2014; 35:429-35. [DOI: 10.1016/j.it.2014.06.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 12/20/2022]
|
42
|
Differential role of nonhomologous end joining factors in the generation, DNA damage response, and myeloid differentiation of human induced pluripotent stem cells. Proc Natl Acad Sci U S A 2014; 111:8889-94. [PMID: 24889605 DOI: 10.1073/pnas.1323649111] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nonhomologous end-joining (NHEJ) is a key pathway for efficient repair of DNA double-strand breaks (DSBs) and V(D)J recombination. NHEJ defects in humans cause immunodeficiency and increased cellular sensitivity to ionizing irradiation (IR) and are variably associated with growth retardation, microcephaly, and neurodevelopmental delay. Repair of DNA DSBs is important for reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). To compare the specific contribution of DNA ligase 4 (LIG4), Artemis, and DNA-protein kinase catalytic subunit (PKcs) in this process and to gain insights into phenotypic variability associated with these disorders, we reprogrammed patient-derived fibroblast cell lines with NHEJ defects. Deficiencies of LIG4 and of DNA-PK catalytic activity, but not Artemis deficiency, were associated with markedly reduced reprogramming efficiency, which could be partially rescued by genetic complementation. Moreover, we identified increased genomic instability in LIG4-deficient iPSCs. Cell cycle synchronization revealed a severe defect of DNA repair and a G0/G1 cell cycle arrest, particularly in LIG4- and DNA-PK catalytically deficient iPSCs. Impaired myeloid differentiation was observed in LIG4-, but not Artemis- or DNA-PK-mutated iPSCs. These results indicate a critical importance of the NHEJ pathway for somatic cell reprogramming, with a major role for LIG4 and DNA-PKcs and a minor, if any, for Artemis.
Collapse
|
43
|
|
44
|
Al-Ghananeem AM, Smith M, Coronel ML, Tran H. Advances in brain targeting and drug delivery of anti-HIV therapeutic agents. Expert Opin Drug Deliv 2013; 10:973-85. [PMID: 23510097 DOI: 10.1517/17425247.2013.781999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Human immunodeficiency virus (HIV) is a neurotropic virus that enters the central nervous system (CNS) early in the course of infection. Although antiretroviral drugs are able to eliminate the majority of the HIV virus in the bloodstream, however, no specific treatment currently exist for CNS infections related to HIV. This is mainly attributed to the poor penetrability of antiretroviral therapy across the blood-brain barrier (BBB), and the protective nature of the BBB. Therefore, in order to increase the efficacy of anti-HIV drugs, novel drug delivery methodologies that can exhibit activity in the CNS are most needed and warranted. AREAS COVERED In this review article, the authors discussed the challenges with delivering drugs to the brain especially under HIV infection pathophysiology status. Also, they discussed the approaches currently being investigated to enhance brain targeting of anti-HIV drugs. A literature search was performed to cover advances in major approaches used to enhance drug delivery to the brain. EXPERT OPINION If drugs could reach the CNS in sufficient quantity by the methodologies discussed, mainly through intranasal administration and the utilization of nanotechnology, this could generate interest in previously abandoned therapeutic agents and enable an entirely novel approach to CNS drug delivery.
Collapse
Affiliation(s)
- Abeer M Al-Ghananeem
- Sullivan University, College of Pharmacy, Department of Pharmaceutical Sciences, 2100 Gardiner Lane West Campus, Louisville, KY 40205, USA.
| | | | | | | |
Collapse
|
45
|
González F, Georgieva D, Vanoli F, Shi ZD, Stadtfeld M, Ludwig T, Jasin M, Huangfu D. Homologous recombination DNA repair genes play a critical role in reprogramming to a pluripotent state. Cell Rep 2013; 3:651-60. [PMID: 23478019 DOI: 10.1016/j.celrep.2013.02.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/15/2013] [Accepted: 02/04/2013] [Indexed: 12/17/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) hold great promise for personalized regenerative medicine. However, recent studies show that iPSC lines carry genetic abnormalities, suggesting that reprogramming may be mutagenic. Here, we show that the ectopic expression of reprogramming factors increases the level of phosphorylated histone H2AX, one of the earliest cellular responses to DNA double-strand breaks (DSBs). Additional mechanistic studies uncover a direct role of the homologous recombination (HR) pathway, a pathway essential for error-free repair of DNA DSBs, in reprogramming. This role is independent of the use of integrative or nonintegrative methods in introducing reprogramming factors, despite the latter being considered a safer approach that circumvents genetic modifications. Finally, deletion of the tumor suppressor p53 rescues the reprogramming phenotype in HR-deficient cells primarily through the restoration of reprogramming-dependent defects in cell proliferation and apoptosis. These mechanistic insights have important implications for the design of safer approaches to creating iPSCs.
Collapse
Affiliation(s)
- Federico González
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Koyama T, Sun B, Tokunaga K, Tatsumi M, Ishizaka Y. DNA damage enhances integration of HIV-1 into macrophages by overcoming integrase inhibition. Retrovirology 2013; 10:21. [PMID: 23432899 PMCID: PMC3605128 DOI: 10.1186/1742-4690-10-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 02/11/2013] [Indexed: 11/10/2022] Open
Abstract
Background The prevention of persistent human immunodeficiency virus type 1 (HIV-1) infection requires the clarification of the mode of viral transduction into resting macrophages. Recently, DNA double-strand breaks (DSBs) were shown to enhance infection by D64A virus, which has a defective integrase catalytic activity (IN-CA). However, the mechanism by which DSBs upregulate viral transduction was unclear. Here we analyzed the roles of DSBs during IN-CA–independent viral transduction into macrophages. Results We used cellular systems with rare-cutting endonucleases and found that D64A virus integrated efficiently into the sites of artificially induced DSBs. This IN-CA-independent viral transduction was blocked by an inhibitor of ataxia telangiectasia mutated protein (ATM) but was resistant to raltegravir (RAL), an inhibitor of integrase activity during strand transfer. Moreover, Vpr, an accessory gene product of HIV-1, induced DSBs in resting macrophages and significantly enhanced the rate of IN-CA-independent viral transduction into macrophages with concomitant production of secondary viruses. Conclusion DSBs contribute to the IN-CA–independent viral infection of macrophages, which is resistant to RAL. Thus, the ATM-dependent cellular pathway and Vpr-induced DNA damage are novel targets for preventing persistent HIV-1 infection.
Collapse
Affiliation(s)
- Takayoshi Koyama
- Department of Intractable Diseases, National Center for Global Health and Medicine, 1-21-1 Toyama, 162-8655, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
47
|
Lin WT, Beattie M, Chen LM, Oktay K, Crawford SL, Gold EB, Cedars M, Rosen M. Comparison of age at natural menopause in BRCA1/2 mutation carriers with a non-clinic-based sample of women in northern California. Cancer 2013; 119:1652-9. [PMID: 23362014 DOI: 10.1002/cncr.27952] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/05/2012] [Accepted: 11/19/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND Germline mutations in BRCA1 and BRCA2 (BRCA1/2) are related to an increased lifetime risk of developing breast and ovarian cancer. Although risk-reducing salpingo-oophorectomy reduces the risk of both cancers, loss of fertility is a major concern. A recent study suggested an association between BRCA1 mutation and occult primary ovarian insufficiency. The objective of the current study was to determine whether BRCA1/2 mutation carriers have an earlier onset of natural menopause compared with unaffected women. METHODS White carriers of the BRCA1/2 gene (n = 382) were identified within the Breast Cancer Risk Program Registry at the University of California at San Francisco and compared with non-clinic-based white women in northern California (n = 765). The 2 groups were compared with regard to median age at the time of natural menopause before and after adjustment for known risk factors, and the role of smoking within each group was examined using the Kaplan-Meier approach for unadjusted analyses and Cox proportional hazards regression analyses for adjusted analyses. RESULTS The median age at the time of natural menopause in the BRCA1/2 carriers was significantly younger than among the unaffected sample (50 years vs 53 years; P < .001). The unadjusted hazard ratio for natural menopause when comparing BRCA1/2 carriers with unaffected women was 4.06 (95% confidence interval, 3.03-5.45) and was 3.98 (95% confidence interval, 2.87-5.53) after adjusting for smoking, parity, and oral contraceptive use. For BRCA1/2 carriers who were current heavy smokers (smoking ≥ 20 cigarettes/day), the median age at natural menopause was 46 years versus 49 years for nonsmokers (P = .027). CONCLUSIONS The BRCA1/2 mutation was associated with a significantly earlier age at natural menopause, and heavy smoking compounded this risk. Because the relationship between menopause and the end of natural fertility is considered to be fixed, these findings suggest the risk of earlier infertility among BRCA1/2 carriers.
Collapse
Affiliation(s)
- Wayne T Lin
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California at San Francisco, San Francisco, California 94122, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
The Intersection of Genetics and Epigenetics: Reactivation of Mammalian LINE-1 Retrotransposons by Environmental Injury. ENVIRONMENTAL EPIGENOMICS IN HEALTH AND DISEASE 2013. [DOI: 10.1007/978-3-642-23380-7_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
49
|
LEDGF (p75) promotes DNA-end resection and homologous recombination. Nat Struct Mol Biol 2012; 19:803-10. [DOI: 10.1038/nsmb.2314] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/24/2012] [Indexed: 12/11/2022]
|
50
|
Doerfler W. Impact of foreign DNA integration on tumor biology and on evolution via epigenetic alterations. Epigenomics 2012; 4:41-9. [PMID: 22332657 DOI: 10.2217/epi.11.111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The insertion of foreign DNA into mammalian genomes can alter their methylation and transcription patterns at remote sites from the locus of foreign DNA integration. The mechanisms leading to these fundamental changes and their frequencies are unknown. Sites and extent of changes in the recipient cells might depend on the location of foreign DNA integration. In the second part of this review, it will be hypothesized that the insertion event itself, for example, of tumor viral DNA via its epigenetic genome-wide consequences, plays an important role in oncogenesis. During evolution, the impact of ancient retrotransposon or retroviral genomes and the ensuing epigenetic alterations in the recipient genomes might have generated cells with completely different transcriptional profiles. Due to the continued presence of the transgenomes these alterations were genetically stable and were selected for or against by the environmental conditions prevalent at the time. These evolutionary effects are very different from those postulated for insertional mutagenesis, added genetic information or regulatory elements placed into the vicinity of cellular functions.
Collapse
Affiliation(s)
- Walter Doerfler
- Institute for Virology, Erlangen University, Erlangen, Germany.
| |
Collapse
|