1
|
Tang X, Lu H, Tarwater PM, Silverberg DL, Schorl C, Ramratnam B. Adeno-Associated Virus (AAV)-Delivered Exosomal TAT and BiTE Molecule CD4-αCD3 Facilitate the Elimination of CD4 T Cells Harboring Latent HIV-1. Microorganisms 2024; 12:1707. [PMID: 39203549 PMCID: PMC11357122 DOI: 10.3390/microorganisms12081707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Combinatorial antiretroviral therapy (cART) has transformed HIV infection from a death sentence to a controllable chronic disease, but cannot eliminate the virus. Latent HIV-1 reservoirs are the major obstacles to cure HIV-1 infection. Previously, we engineered exosomal Tat (Exo-Tat) to reactivate latent HIV-1 from the reservoir of resting CD4+ T cells. Here, we present an HIV-1 eradication platform, which uses our previously described Exo-Tat to activate latent virus from resting CD4+ T cells guided by the specific binding domain of CD4 in interleukin 16 (IL16), attached to the N-terminus of exosome surface protein lysosome-associated membrane protein 2 variant B (Lamp2B). Cells with HIV-1 surface protein gp120 expressed on the cell membranes are then targeted for immune cytolysis by a BiTE molecule CD4-αCD3, which colocalizes the gp120 surface protein of HIV-1 and the CD3 of cytotoxic T lymphocytes. Using primary blood cells obtained from antiretroviral treated individuals, we find that this combined approach led to a significant reduction in replication-competent HIV-1 in infected CD4+ T cells in a clonal in vitro cell system. Furthermore, adeno-associated virus serotype DJ (AAV-DJ) was used to deliver Exo-Tat, IL16lamp2b and CD4-αCD3 genes by constructing them in one AAV-DJ vector (the plasmid was named pEliminator). The coculture of T cells from HIV-1 patients with Huh-7 cells infected with AAV-Eliminator viruses led to the clearance of HIV-1 reservoir cells in the in vitro experiment, which could have implications for reducing the viral reservoir in vivo, indicating that Eliminator AAV viruses have the potential to be developed into therapeutic biologics to cure HIV-1 infection.
Collapse
Affiliation(s)
- Xiaoli Tang
- Division of Infectious Diseases, Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (X.T.); (H.L.)
| | - Huafei Lu
- Division of Infectious Diseases, Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (X.T.); (H.L.)
| | - Patrick M. Tarwater
- Department of Epidemiology and Biostatistics, Texas A&M School of Public Health, College Station, TX 77843, USA;
| | - David L. Silverberg
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI 02906, USA;
| | - Christoph Schorl
- The Brown University Genomics Core, Providence, RI 02906, USA;
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02906, USA
| | - Bharat Ramratnam
- Division of Infectious Diseases, Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (X.T.); (H.L.)
- COBRE Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI 02903, USA
- Clinical Research Center of Lifespan, Providence, RI 02903, USA
| |
Collapse
|
2
|
Akade E, Jalilian S. The role of high mobility group AT-hook 1 in viral infections: Implications for cancer pathogenesis. Int J Biochem Cell Biol 2024; 169:106532. [PMID: 38278412 DOI: 10.1016/j.biocel.2024.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/25/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
The crucial role of high mobility group AT-hook 1 (HMGA1) proteins in nuclear processes such as gene transcription, DNA replication, and chromatin remodeling is undeniable. Elevated levels of HMGA1 have been associated with unfavorable clinical outcomes and adverse differentiation status across various cancer types. HMGA1 regulates a diverse array of biological pathways, including tumor necrosis factor-alpha/nuclear factor-kappa B (TNF-α/NF-κB), epidermal growth factor receptor (EGFR), Hippo, Rat sarcoma/extracellular signal-regulated kinase (Ras/ERK), protein kinase B (Akt), wingless-related integration site/beta-catenin (Wnt/beta-catenin), and phosphoinositide 3-kinase/protein kinase B (PI3-K/Akt). While researchers have extensively investigated tumors in the reproductive, digestive, urinary, and hematopoietic systems, mounting evidence suggests that HMGA1 plays a critical role as a tumorigenic factor in tumors across all functional systems. Given its broad interaction network, HMGA1 is an attractive target for viral manipulation. Some viruses, including herpes simplex virus type 1, human herpesvirus 8, human papillomavirus, JC virus, hepatitis B virus, human immunodeficiency virus type 1, severe acute respiratory syndrome Coronavirus 2, and influenza viruses, utilize HMGA1 influence for infection. This interaction, particularly in oncogenesis, is crucial. Apart from the direct oncogenic effect of some of the mentioned viruses, the hit-and-run theory postulates that viruses can instigate cancer even before being completely eradicated from the host cell, implying a potentially greater impact of viruses on cancer development than previously assumed. This review explores the interplay between HMGA1, viruses, and host cellular machinery, aiming to contribute to a deeper understanding of viral-induced oncogenesis, paving the way for innovative strategies in cancer research and treatment.
Collapse
Affiliation(s)
- Esma'il Akade
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahram Jalilian
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Beliakova-Bethell N. Targeting noncoding RNAs to reactivate or eliminate latent HIV reservoirs. Curr Opin HIV AIDS 2024; 19:47-55. [PMID: 38169367 PMCID: PMC10872953 DOI: 10.1097/coh.0000000000000838] [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] [Indexed: 01/05/2024]
Abstract
PURPOSE OF REVIEW Expression of noncoding RNAs (ncRNAs) is more tissue and cell type-specific than expression of protein-coding genes. Understanding the mechanisms of action of ncRNAs and their roles in HIV replication and latency may inform targets for the latent HIV reservoir reactivation or elimination with high specificity to CD4 + T cells latently infected with HIV. RECENT FINDINGS While the number of studies in the field of ncRNAs and HIV is limited, evidence points to complex interactions between different ncRNAs, protein-coding RNAs, and proteins. Latency-reversing agents modulate the expression of ncRNAs, with some effects being inhibitory for HIV reactivation. An important limitation of basic research on the ncRNA mechanisms of action is the reliance on cell lines. Because of cell type specificity, it is uncertain whether the ncRNAs function similarly in primary cells. SUMMARY Comprehensive functional screens to uncover all ncRNAs that regulate HIV expression and the detailed exploration of their mechanisms of action in relevant cell types are needed to identify promising targets for HIV reservoir clearance. Classes of ncRNAs as a whole rather than individual ncRNAs might represent an attractive target for reservoir elimination. Compound screens for latency reversal should factor in the complexity of their effects on ncRNAs.
Collapse
Affiliation(s)
- Nadejda Beliakova-Bethell
- Department of Medicine, University of California at San Diego, CA, USA
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, USA
| |
Collapse
|
4
|
Beauchemin R, Merindol N, Fantino E, Lavoie P, Nouemssi SB, Meddeb-Mouelhi F, Desgagné-Penix I. Successful reversal of transgene silencing in Chlamydomonas reinhardtii. Biotechnol J 2024; 19:e2300232. [PMID: 37975165 DOI: 10.1002/biot.202300232] [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: 05/23/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Chlamydomonas reinhardtii has been successfully engineered to produce compounds of interest following transgene integration and heterologous protein expression. The advantages of this model include the availability of validated tools for bioengineering, its photosynthetic ability, and its potential use as biofuel. Despite this, breakthroughs have been hindered by its ability to silence transgene expression through epigenetic changes. Histone deacetylases (HDAC) are main players in gene expression. We hypothesized that transgene silencing can be reverted with chemical treatments using HDAC inhibitors. To analyze this, we transformed C. reinhardtii, integrating into its genome the mVenus reporter gene under the HSP70-rbcs2 promoter. From 384 transformed clones, 88 (22.9%) displayed mVenus positive (mVenus+ ) cells upon flow-cytometry analysis. Five clones with different fluorescence intensities were selected. The number of integrated copies was measured by qPCR. Transgene expression levels were followed over the growth cycle and upon SAHA treatment, using a microplate reader, flow cytometry, RT-qPCR, and western blot analysis. First, we observed that expression varies with the cell cycle, reaching a maximum level just before the stationary phase in all clones. Second, we uncovered that supplementation with HDAC inhibitors of the hydroxamate family, such as vorinostat (suberoylanilide-hydroxamic-acid, SAHA) at the initiation of culture increases the frequency (% of mVenus+ cells) and the level of transgene expression per cell over the whole growth cycle, through histone deacetylase inhibition. Thus, we propose a new tool to successfully trigger the expression of heterologous proteins in the green algae C. reinhardtii, overcoming its main obstacle as an expression platform.
Collapse
Affiliation(s)
- Rémy Beauchemin
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Natacha Merindol
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Elisa Fantino
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Pamela Lavoie
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Serge Basile Nouemssi
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Fatma Meddeb-Mouelhi
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Isabel Desgagné-Penix
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
- Plant Biology Research Group, Trois-Rivières, Québec, Canada
| |
Collapse
|
5
|
Zhang X, Qazi AA, Deshmukh S, Lobato Ventura R, Mukim A, Beliakova-Bethell N. Single-cell RNA sequencing reveals common and unique gene expression profiles in primary CD4+ T cells latently infected with HIV under different conditions. Front Cell Infect Microbiol 2023; 13:1286168. [PMID: 38156317 PMCID: PMC10754520 DOI: 10.3389/fcimb.2023.1286168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Background The latent HIV reservoir represents the major barrier to a cure. One curative strategy is targeting diseased cells for elimination based on biomarkers that uniquely define these cells. Single-cell RNA sequencing (scRNA-seq) has enabled the identification of gene expression profiles associated with disease at the single-cell level. Because HIV provirus in many cells during latency is not entirely silent, it became possible to determine gene expression patterns in a subset of cells latently infected with HIV. Objective The primary objective of this study was the identification of the gene expression profiles of single latently infected CD4+ T cells using scRNA-seq. Different conditions of latency establishment were considered. The identified profiles were then explored to prioritize the identified genes for future experimental validation. Methods To facilitate gene prioritization, three approaches were used. First, we characterized and compared the gene expression profiles of HIV latency established in different environments: in cells that encountered an activation stimulus and then returned to quiescence, and in resting cells that were infected directly via cell-to-cell viral transmission from autologous activated, productively infected cells. Second, we characterized and compared the gene expression profiles of HIV latency established with viruses of different tropisms, using an isogenic pair of CXCR4- and CCR5-tropic viruses. Lastly, we used proviral expression patterns in cells from people with HIV to more accurately define the latently infected cells in vitro. Results Our analyses demonstrated that a subset of genes is expressed differentially between latently infected and uninfected cells consistently under most conditions tested, including cells from people with HIV. Our second important observation was the presence of latency signatures, associated with variable conditions when latency was established, including cellular exposure and responsiveness to a T cell receptor stimulus and the tropism of the infecting virus. Conclusion Common signatures, specifically genes that encode proteins localized to the cell surface, should be prioritized for further testing at the protein level as biomarkers for the ability to enrich or target latently infected cells. Cell- and tropism-dependent biomarkers may need to be considered in developing targeting strategies to ensure that all the different reservoir subsets are eliminated.
Collapse
Affiliation(s)
- Xinlian Zhang
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA, United States
| | - Andrew A. Qazi
- Veterans Affairs (VA), San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, United States
| | - Savitha Deshmukh
- Veterans Affairs (VA), San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, United States
| | - Roni Lobato Ventura
- Veterans Affairs (VA), San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, United States
| | - Amey Mukim
- Veterans Affairs (VA), San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, United States
| | - Nadejda Beliakova-Bethell
- Veterans Affairs (VA), San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
6
|
Beliakova-Bethell N, Manousopoulou A, Deshmukh S, Mukim A, Richman DD, Garbis SD, Spina CA. Integrated proteomics and transcriptomics analyses identify novel cell surface markers of HIV latency. Virology 2022; 573:50-58. [PMID: 35714458 PMCID: PMC10427345 DOI: 10.1016/j.virol.2022.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Elimination of the latent HIV cell reservoir may be possible, if the molecular identity of latently infected cells were fully elucidated. We conducted comprehensive molecular profiling, at the protein and RNA levels, of primary T cells latently infected with HIV in vitro. Isobaric labelling quantitative proteomics and RNA sequencing identified 1453 proteins and 618 genes, altered in latently infected cells compared to mock-infected controls (p < 0.05). Biomarker selection was based on results from integrated data analysis. Relative enrichment for latently infected cells was monitored using flow cytometric sorting and the HIV integrant assay. Antibodies against selected proteins, encoded by CEACAM1 and PLXNB2, enabled enrichment of latently infected cells from cell mixtures by 3-10 fold (5.8 average, p < 0.001), comparable to levels obtained with biomarkers reported previously. Individual biomarkers are likely linked to subsets of latently infected cells, and an extended antibody panel will be required to inclusively target the latent HIV reservoir.
Collapse
Affiliation(s)
- Nadejda Beliakova-Bethell
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, USA; University of California at San Diego, CA, USA.
| | - Antigoni Manousopoulou
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK; Proteas Bioanalytics Inc., BioLabs at the Lundquist Institute, Torrance, CA, USA
| | | | - Amey Mukim
- University of California at San Diego, CA, USA
| | - Douglas D Richman
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, USA; University of California at San Diego, CA, USA
| | - Spiros D Garbis
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK; Proteas Bioanalytics Inc., BioLabs at the Lundquist Institute, Torrance, CA, USA
| | - Celsa A Spina
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, CA, USA; University of California at San Diego, CA, USA
| |
Collapse
|
7
|
Salahong T, Schwartz C, Sungthong R. Are BET Inhibitors yet Promising Latency-Reversing Agents for HIV-1 Reactivation in AIDS Therapy? Viruses 2021; 13:v13061026. [PMID: 34072421 PMCID: PMC8228869 DOI: 10.3390/v13061026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
AIDS first emerged decades ago; however, its cure, i.e., eliminating all virus sources, is still unachievable. A critical burden of AIDS therapy is the evasive nature of HIV-1 in face of host immune responses, the so-called "latency." Recently, a promising approach, the "Shock and Kill" strategy, was proposed to eliminate latently HIV-1-infected cell reservoirs. The "Shock and Kill" concept involves two crucial steps: HIV-1 reactivation from its latency stage using a latency-reversing agent (LRA) followed by host immune responses to destroy HIV-1-infected cells in combination with reinforced antiretroviral therapy to kill the progeny virus. Hence, a key challenge is to search for optimal LRAs. Looking at epigenetics of HIV-1 infection, researchers proved that some bromodomains and extra-terminal motif protein inhibitors (BETis) are able to reactivate HIV-1 from latency. However, to date, only a few BETis have shown HIV-1-reactivating functions, and none of them have yet been approved for clinical trial. In this review, we aim to demonstrate the epigenetic roles of BETis in HIV-1 infection and HIV-1-related immune responses. Possible future applications of BETis and their HIV-1-reactivating properties are summarized and discussed.
Collapse
Affiliation(s)
- Thanarat Salahong
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Christian Schwartz
- Research Unit 7292, DHPI, IUT Louis Pasteur, University of Strasbourg, 67300 Schiltigheim, France
- Correspondence: (C.S.); (R.S.)
| | - Rungroch Sungthong
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
- Laboratory of Hydrology and Geochemistry of Strasbourg, University of Strasbourg, UMR 7517 CNRS/EOST, 67084 Strasbourg CEDEX, France
- Correspondence: (C.S.); (R.S.)
| |
Collapse
|
8
|
Heinson AI, Woo J, Mukim A, White CH, Moesker B, Bosque A, Spina CA, Woelk CH, Macarthur BD, Beliakova-Bethell N. Micro RNA Targets in HIV Latency: Insights into Novel Layers of Latency Control. AIDS Res Hum Retroviruses 2021; 37:109-121. [PMID: 33045840 PMCID: PMC7876363 DOI: 10.1089/aid.2020.0150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite the considerable progress that has been made in identifying cellular factors and pathways that contribute to establishment and maintenance of the latent HIV reservoir, it remains the major obstacle to eradicating this virus. Most recently, noncoding genes have been implicated in regulation of HIV expression. In this study, small RNA sequencing was used to profile expression of microRNAs (miRNAs) in a primary CD4+ T cell in vitro model of HIV latency. Previously, we have shown that protein-coding genes dysregulated in this model were enriched for the p53 signaling pathway, which was confirmed experimentally. We further found a link between p53 signaling and dysregulated long noncoding RNAs. In this study, we hypothesized that miRNAs may provide an additional level of regulation of the p53 signaling pathway during HIV latency. Twenty-six miRNAs were identified to be dysregulated in our latency model. A subset of these miRNAs was validated by real-time quantitative polymerase chain reaction. Predicted messenger RNA (mRNA) targets and cellular pathways enriched for mRNA targets were identified using several analytical methods. Our analyses showed that many protein-coding genes and pathways targeted by dysregulated miRNAs have relevance to regulation of HIV expression or establishment of HIV latency. The p53 signaling pathway was found among pathways that were targeted by dysregulated miRNAs at a greater level than expected by chance. This study provides a mechanistic insight into regulation of the p53 pathway through miRNAs that may contribute to the establishment of latency.
Collapse
Affiliation(s)
- Ashley I. Heinson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jeongmin Woo
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Amey Mukim
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California, USA
| | - Cory H. White
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Bastiaan Moesker
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Alberto Bosque
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, District of Columbia, USA
| | - Celsa A. Spina
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California, USA
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | | | - Ben D. Macarthur
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nadejda Beliakova-Bethell
- VA San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
9
|
Grabowska K, Harwood E, Ciborowski P. HIV and Proteomics: What We Have Learned from High Throughput Studies. Proteomics Clin Appl 2021; 15:e2000040. [PMID: 32978881 PMCID: PMC7900993 DOI: 10.1002/prca.202000040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/04/2020] [Indexed: 12/17/2022]
Abstract
The accelerated development of technology over the last three decades has driven biological sciences to high-throughput profiling experiments, now broadly referred to as systems biology. The unprecedented improvement of analytical instrumentation has opened new avenues for more complex experimental designs and expands the knowledge in genomics, proteomics, and other omics fields. Despite the collective efforts of hundreds of researchers, gleaning all the expected information from omics experiments is still quite far. This paper summarizes what has been learned from high-throughput proteomics studies thus far, and what is believed should be done to reveal even more valuable information from such studies. It is drawn from the background in using proteomics to study human immunodeficiency virus 1 infection of macrophages and/or T cells, but it is believed that some conclusions will be more broadly applicable.
Collapse
Affiliation(s)
- Kinga Grabowska
- Laboratory of Virus Molecular BiologyIntercollegiate Faculty of BiotechnologyUniversity of GdanskGdansk80‐307Poland
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| | - Emma Harwood
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| |
Collapse
|
10
|
Fujinaga K, Cary DC. Experimental Systems for Measuring HIV Latency and Reactivation. Viruses 2020; 12:v12111279. [PMID: 33182414 PMCID: PMC7696534 DOI: 10.3390/v12111279] [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: 08/15/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
The final obstacle to achieving a cure to HIV/AIDS is the presence of latent HIV reservoirs scattered throughout the body. Although antiretroviral therapy maintains plasma viral loads below the levels of detection, upon cessation of therapy, the latent reservoir immediately produces infectious progeny viruses. This results in elevated plasma viremia, which leads to clinical progression to AIDS. Thus, if a HIV cure is ever to become a reality, it will be necessary to target and eliminate the latent reservoir. To this end, tremendous effort has been dedicated to locate the viral reservoir, understand the mechanisms contributing to latency, find optimal methods to reactivate HIV, and specifically kill latently infected cells. Although we have not yet identified a therapeutic approach to completely eliminate HIV from patients, these efforts have provided many technological breakthroughs in understanding the underlying mechanisms that regulate HIV latency and reactivation in vitro. In this review, we summarize and compare experimental systems which are frequently used to study HIV latency. While none of these models are a perfect proxy for the complex systems at work in HIV+ patients, each aim to replicate HIV latency in vitro.
Collapse
Affiliation(s)
- Koh Fujinaga
- Division of Rheumatology, Department of Medicine, School of Medicine, University of California, San Francisco, CA 94143-0703, USA
- Correspondence: ; Tel.: +1-415-502-1908
| | - Daniele C. Cary
- Department of Medicine, Microbiology, and Immunology, School of Medicine, University of California, San Francisco, CA 94143-0703, USA;
| |
Collapse
|
11
|
Cellular Gene Modulation of HIV-Infected CD4 T Cells in Response to Serial Treatment with the Histone Deacetylase Inhibitor Vorinostat. J Virol 2020; 94:JVI.00351-20. [PMID: 32295913 PMCID: PMC7307144 DOI: 10.1128/jvi.00351-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase inhibitors are widely studied HIV latency-reversing agents (LRAs). VOR, an HDACi, induces histone acetylation and chromatin remodeling and modulates host and HIV gene expression. However, the relationship between these events is poorly defined, and clinical studies suggest diminished HIV reactivation in resting CD4 T cells with daily exposure to VOR. Our study provides evidence that VOR induces a consistent level of host cell gene transcription following intermittent exposure. In addition, in response to VOR exposure a gene signature that was conserved across single and serial exposures both in vitro and in vivo was identified, indicating that VOR can consistently and reproducibly modulate transcriptional host responses. However, as the HIV response to HDACi declines over time, other factors modulate viral reactivation in vivo despite robust HDAC activity. The identified host gene VOR biomarkers can be used for monitoring the pharmacodynamic activity of HDAC inhibitors. Histone deacetylase inhibitors (HDACi) are the most widely studied HIV latency-reversing agents (LRAs). The HDACi suberoylanilide hydroxamic acid (vorinostat [VOR]) has been employed in several clinical HIV latency reversal studies, as well as in vitro models of HIV latency, and has been shown to effectively induce HIV RNA and protein expression. Despite these findings, response to HDACi can vary, particularly with intermittent dosing, and information is lacking on the relationship between the host transcriptional response and HIV latency reversal. Here, we report on global gene expression responses to VOR and examine the longevity of the transcriptional response in various cellular models. We found that many genes are modulated at 6 h post-VOR treatment in HCT116, Jurkat, and primary resting CD4 T cells, yet return to baseline levels after an 18-h VOR-free period. With repeat exposure to VOR in resting CD4 T cells, we found similar and consistent transcriptional changes at 6 h following each serial treatment. In addition, serial exposure in HIV-infected suppressed donor CD4 T cells showed consistent transcriptional changes after each exposure to VOR. We identified five host genes that were strongly and consistently modulated following histone deacetylase (HDAC) inhibition; three (H1F0, IRGM, and WIPI49) were upregulated, and two (PHF15 and PRDM10) were downregulated. These genes demonstrated consistent modulation in peripheral blood mononuclear cell (PBMC) samples from HIV-positive (HIV+) participants who received either single or multiple doses of 400 mg of VOR. Interestingly, the host transcriptional response did not predict induction of cell-associated HIV RNA, suggesting that other cellular factors play key roles in HIV latency reversal in vivo despite robust HDACi pharmacological activity. IMPORTANCE Histone deacetylase inhibitors are widely studied HIV latency-reversing agents (LRAs). VOR, an HDACi, induces histone acetylation and chromatin remodeling and modulates host and HIV gene expression. However, the relationship between these events is poorly defined, and clinical studies suggest diminished HIV reactivation in resting CD4 T cells with daily exposure to VOR. Our study provides evidence that VOR induces a consistent level of host cell gene transcription following intermittent exposure. In addition, in response to VOR exposure a gene signature that was conserved across single and serial exposures both in vitro and in vivo was identified, indicating that VOR can consistently and reproducibly modulate transcriptional host responses. However, as the HIV response to HDACi declines over time, other factors modulate viral reactivation in vivo despite robust HDAC activity. The identified host gene VOR biomarkers can be used for monitoring the pharmacodynamic activity of HDAC inhibitors.
Collapse
|
12
|
Abewe H, Deshmukh S, Mukim A, Beliakova-Bethell N. Use of GapmeRs for gene expression knockdowns in human primary resting CD4+ T cells. J Immunol Methods 2020; 476:112674. [PMID: 31629740 PMCID: PMC6939142 DOI: 10.1016/j.jim.2019.112674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022]
Abstract
Human primary resting CD4+ T cells are difficult to transfect while preserving viability. The present study evaluated gymnotic delivery and RNase H1-dependent gene expression knockdown mediated by antisense oligonucleotides, called GapmeRs. Exposure of primary resting CD4+ T cells to GapmeRs did not cause cell activation or affect cell viability. Gene expression knockdowns were stable at least up to 48 h after removal of GapmeRs from culture. Exposure to GapmeRs resulted in comparable levels of degradation along the entire transcript, which could be important when studying function of regulatory long non-coding RNAs. Efficiency of transcript degradation was not solely dependent on the dose of GapmeR, RNA target and its localization. When using GapmeRs, some optimization is required, and all targets have to be individually tested; however, using GapmeRs is advantageous in experiments where preservation of the resting state of the human primary CD4+ T cells and targeting nuclear RNAs are desired. In certain cases, combining GapmeR with siRNA for the same target may improve knockdown efficiency.
Collapse
Affiliation(s)
- Hosiana Abewe
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA; University of California San Diego, La Jolla, CA 92093, USA
| | - Savitha Deshmukh
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA
| | - Amey Mukim
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA
| | - Nadejda Beliakova-Bethell
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA; University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Trypsteen W, White CH, Mukim A, Spina CA, De Spiegelaere W, Lefever S, Planelles V, Bosque A, Woelk CH, Vandekerckhove L, Beliakova-Bethell N. Long non-coding RNAs and latent HIV - A search for novel targets for latency reversal. PLoS One 2019; 14:e0224879. [PMID: 31710657 PMCID: PMC6844474 DOI: 10.1371/journal.pone.0224879] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/23/2019] [Indexed: 12/23/2022] Open
Abstract
The latent cellular reservoir of HIV is recognized as the major barrier to cure from HIV infection. Long non-coding RNAs (lncRNAs) are more tissue and cell type-specific than protein coding genes, and may represent targets of choice for HIV latency reversal. Using two in vitro primary T-cell models, we identified lncRNAs dysregulated in latency. PVT1 and RP11-347C18.3 were up-regulated in common between the two models, and RP11-539L10.2 was down-regulated. The major component of the latent HIV reservoir, memory CD4+ T-cells, had higher expression of these lncRNAs, compared to naïve T-cells. Guilt-by-association analysis demonstrated that lncRNAs dysregulated in latency were associated with several cellular pathways implicated in HIV latency establishment and maintenance: proteasome, spliceosome, p53 signaling, and mammalian target of rapamycin (MTOR). PVT1, RP11-347C18.3, and RP11-539L10.2 were down-regulated by latency reversing agents, suberoylanilide hydroxamic acid and Romidepsin, suggesting that modulation of lncRNAs is a possible secondary mechanism of action of these compounds. These results will facilitate prioritization of lncRNAs for evaluation as targets for HIV latency reversal. Importantly, our study provides insights into regulatory function of lncRNA during latent HIV infection.
Collapse
Affiliation(s)
- Wim Trypsteen
- HIV Cure Research Center, Department of Internal Medicine, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Cory H. White
- Faculty of Medicine, University of Southampton, Southampton, Hants, United Kingdom
| | - Amey Mukim
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States of America
| | - Celsa A. Spina
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States of America
- Department of Pathology, University of California San Diego, La Jolla, CA, United States of America
| | - Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Sciences, Ghent University, Ghent, Belgium
| | - Steve Lefever
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Vicente Planelles
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Alberto Bosque
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States of America
| | - Christopher H. Woelk
- Faculty of Medicine, University of Southampton, Southampton, Hants, United Kingdom
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Nadejda Beliakova-Bethell
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States of America
- Department of Medicine, University of California San Diego, La Jolla, CA, United States of America
| |
Collapse
|
14
|
Beliakova-Bethell N, Mukim A, White CH, Deshmukh S, Abewe H, Richman DD, Spina CA. Histone deacetylase inhibitors induce complex host responses that contribute to differential potencies of these compounds in HIV reactivation. J Biol Chem 2019; 294:5576-5589. [PMID: 30745362 PMCID: PMC6462528 DOI: 10.1074/jbc.ra118.005185] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/07/2019] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors (HDACis) have been widely tested in clinical trials for their ability to reverse HIV latency but have yielded only limited success. One HDACi, suberoylanilide hydroxamic acid (SAHA), exhibits off-target effects on host gene expression predicted to interfere with induction of HIV transcription. Romidepsin (RMD) has higher potency and specificity for class I HDACs implicated in maintaining HIV provirus in the latent state. More robust HIV reactivation has indeed been achieved with RMD use ex vivo than with SAHA; however, reduction of viral reservoir size has not been observed in clinical trials. Therefore, using RNA-Seq, we sought to compare the effects of SAHA and RMD on gene expression in primary CD4+ T cells. Among the genes whose expression was modulated by both HDACi agents, we identified genes previously implicated in HIV latency. Two genes, SMARCB1 and PARP1, whose modulation by SAHA and RMD is predicted to inhibit HIV reactivation, were evaluated in the major maturation subsets of CD4+ T cells and were consistently either up- or down-regulated by both HDACi compounds. Our results indicate that despite having different potencies and HDAC specificities, SAHA and RMD modulate an overlapping set of genes, implicated in HIV latency regulation. Some of these genes merit exploration as additional targets to improve the therapeutic outcomes of "shock and kill" strategies. The overall complexity of HDACi-induced responses among host genes with predicted stimulatory or inhibitory effects on HIV expression likely contributes to differential HDACi potencies and dictates the outcome of HIV reactivation.
Collapse
Affiliation(s)
- Nadejda Beliakova-Bethell
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
- the University of California San Diego, La Jolla, California 92093
| | - Amey Mukim
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
| | - Cory H White
- the University of California San Diego, La Jolla, California 92093
| | - Savitha Deshmukh
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
| | - Hosiana Abewe
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
- the University of California San Diego, La Jolla, California 92093
| | - Douglas D Richman
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
- the University of California San Diego, La Jolla, California 92093
| | - Celsa A Spina
- From the Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California 92161 and
- the University of California San Diego, La Jolla, California 92093
| |
Collapse
|
15
|
Franci G, Folliero V, Cammarota M, Zannella C, Sarno F, Schiraldi C, de Lera AR, Altucci L, Galdiero M. Epigenetic modulator UVI5008 inhibits MRSA by interfering with bacterial gyrase. Sci Rep 2018; 8:13117. [PMID: 30177735 PMCID: PMC6120918 DOI: 10.1038/s41598-018-31135-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023] Open
Abstract
The impact of multi-drug resistant bacterial strains on human health is reaching worrisome levels. Over 2 million people are infected by resistant bacteria, and more than 700,000 people die each year because of the continuous spread of resistant strains. The development of new antibiotics and the prudent use of existing ones to prolong their lifespan require a constant effort by drug industries and healthcare workers. The re-purposing of existing drugs for use as antimicrobial agents would streamline the development of new antibacterial strategies. As part of this effort, we screened a panel of drugs previously characterized to be epigenetic modulators/pro-apoptotic/differentiative drugs. We selected a few compounds that alter Gram-positive growth. Among these, UVI5008, a derivative of the natural compound psammaplin A (Psa_A), was identified. The interaction of Psa_A with the DNA gyrase enzyme has been shown, and here, we hypothesized and confirmed the gyrase-specific activity by biochemical assays. UVI5008 exhibited growth inhibition activity against Staphylococcus aureus via structural modification of the cell wall, which was observed by SEM electron microscopy. Based on our findings, we propose UVI5008 as an alternative antibacterial compound against methicillin-resistant (Met.R) S. aureus strains.
Collapse
Affiliation(s)
- Gianluigi Franci
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Veronica Folliero
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Marcella Cammarota
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Carla Zannella
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Federica Sarno
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Chiara Schiraldi
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Angel R de Lera
- Departamento de Química Orgánica, Facultade de Química, Universidade de Vigo, CINBIO and IBIV, Vigo, Spain
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy.
| | - Massimiliano Galdiero
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy.
| |
Collapse
|
16
|
White CH, Beliakova-Bethell N, Lada SM, Breen MS, Hurst TP, Spina CA, Richman DD, Frater J, Magiorkinis G, Woelk CH. Transcriptional Modulation of Human Endogenous Retroviruses in Primary CD4+ T Cells Following Vorinostat Treatment. Front Immunol 2018; 9:603. [PMID: 29706951 PMCID: PMC5906534 DOI: 10.3389/fimmu.2018.00603] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
The greatest obstacle to a cure for HIV is the provirus that integrates into the genome of the infected cell and persists despite antiretroviral therapy. A "shock and kill" approach has been proposed as a strategy for an HIV cure whereby drugs and compounds referred to as latency-reversing agents (LRAs) are used to "shock" the silent provirus into active replication to permit "killing" by virus-induced pathology or immune recognition. The LRA most utilized to date in clinical trials has been the histone deacetylase (HDAC) inhibitor-vorinostat. Potentially, pathological off-target effects of vorinostat may result from the activation of human endogenous retroviruses (HERVs), which share common ancestry with exogenous retroviruses including HIV. To explore the effects of HDAC inhibition on HERV transcription, an unbiased pharmacogenomics approach (total RNA-Seq) was used to evaluate HERV expression following the exposure of primary CD4+ T cells to a high dose of vorinostat. Over 2,000 individual HERV elements were found to be significantly modulated by vorinostat, whereby elements belonging to the ERVL family (e.g., LTR16C and LTR33) were predominantly downregulated, in contrast to LTR12 elements of the HERV-9 family, which exhibited the greatest signal, with the upregulation of 140 distinct elements. The modulation of three different LTR12 elements by vorinostat was confirmed by droplet digital PCR along a dose-response curve. The monitoring of LTR12 expression during clinical trials with vorinostat may be indicated to assess the impact of this HERV on the human genome and host immunity.
Collapse
Affiliation(s)
- Cory H. White
- Faculty of Medicine, University of Southampton, Southampton, Hants, United Kingdom
| | - Nadejda Beliakova-Bethell
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Steven M. Lada
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States
| | - Michael S. Breen
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tara P. Hurst
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Celsa A. Spina
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States
- Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | - Douglas D. Richman
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | - John Frater
- Nuffield Department of Clinical Medicine, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, United Kingdom
| | | | - Christopher H. Woelk
- Faculty of Medicine, University of Southampton, Southampton, Hants, United Kingdom
| |
Collapse
|
17
|
Lu ZX, Xu WJ, Wu YS, Li CY, Chen YT. Identification of Potential Therapeutic Targets in the Liver of Pioglitazone-Treated Type 2 Diabetes Sprague-Dawley Rats via Expression Profile Chip and iTRAQ Assay. J Diabetes Res 2018; 2018:8120847. [PMID: 29744368 PMCID: PMC5878868 DOI: 10.1155/2018/8120847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/14/2018] [Indexed: 11/18/2022] Open
Abstract
The aim of the present study was to identify key antidiabetic nodes in the livers of pioglitazone-treated type 2 diabetes mellitus Sprague-Dawley rats by transcriptomic and proteomic analysis. Rats were randomly divided into the control, the diabetes model, and the pioglitazone-treated groups. After treatment with pioglitazone for 11 weeks, the effects on fasting blood glucose, body weight, and blood biochemistry parameters were evaluated. Microarray and iTRAQ analysis were used to determine the differentially expressed genes/proteins in rat livers. 1.5-fold changes in gene expression and 1.2-fold changes in protein were set as the screening criteria. After treatment with pioglitazone for 11 weeks, fasting blood glucose in pioglitazone-treated rats was significantly lower than that in the model group. There was a tendency for pioglitazone to reduce TC, TG, TP, ALB, BUN, and HDL-c levels. Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) were applied to analyze differentially expressed genes/proteins. Furthermore, Western blotting and RT-qPCR were used to validate the results of microarray and iTRAQ. In conclusion, Cyp7a1, Cp, and RT1-EC2 are differentially expressed genes/proteins since they showed a similar trend in rats in the model group and the pioglitazone-treated group.
Collapse
Affiliation(s)
- Zhong-Xia Lu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wen-Jun Xu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yang-Sheng Wu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chang-Yu Li
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Tao Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
18
|
Datta PK, Kaminski R, Hu W, Pirrone V, Sullivan NT, Nonnemacher MR, Dampier W, Wigdahl B, Khalili K. HIV-1 Latency and Eradication: Past, Present and Future. Curr HIV Res 2017; 14:431-441. [PMID: 27009094 DOI: 10.2174/1570162x14666160324125536] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/04/2015] [Accepted: 01/16/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND It is well established that antiretroviral therapy (ART), while highly effective in controlling HIV replication, cannot eliminate virus from the body. Therefore, the majority of HIV-1-infected individuals remain at risk for developing AIDS due to persistence of infected reservoir cells serving as a source of virus re-emergence. Several reservoirs containing replication competent HIV-1 have been identified, most notably CD4+ T cells. Cells of the myeloid lineage, which are the first line of defense against pathogens and participate in HIV dissemination into sanctuary organs, also serve as cellular reservoirs of HIV-1. In latently infected resting CD4+ T cells, the integrated copies of proviral DNA remain in a dormant state, yet possess the ability to produce replication competent virus after cellular activation. Studies have demonstrated that modification of chromatin structure plays a role in establishing persistence, in part suggesting that latency is, controlled epigenetically. CONCLUSION Current efforts to eradicate HIV-1 from this cell population focus primarily on a &quot;shock and kill&quot; approach through cellular reactivation to trigger elimination of virus producing cells by cytolysis or host immune responses. However, studies revealed several limitations to this approach that require more investigation to assess its clinical application. Recent advances in gene editing technology prompted use of this approach for inactivating integrated proviral DNA in the genome of latently infected cells. This technology, which requires a detailed understanding of the viral genetics and robust delivery, may serve as a powerful strategy to eliminate the latent reservoir in the host leading to a sterile cure of AIDS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, 7th Floor, Philadelphia, PA 19140, USA.
| |
Collapse
|
19
|
Yang Q, Ding Y, Feng F, Pan E, Fan X, Ma X, Chen L, Zhao J, Sun C. Structure-optimized dihydropyranoindole derivative GIBH-LRA002 potentially reactivated viral latency in primary CD4+ T lymphocytes of chronic HIV-1 patients. MEDCHEMCOMM 2017; 8:1806-1809. [PMID: 30108890 DOI: 10.1039/c7md00327g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/21/2017] [Indexed: 12/29/2022]
Abstract
Based on structure modification and a high-throughput Jurkat-Lat cell screening model, we found that GIBH-LRA002, ethyl-2-amino-3-cyano-9-methyl-4-(trifluoromethyl)-4,9-dihydropyrano[2,3-b]indole-4-carboxylate, effectively reactivated the latent proviruses in a Jurkat-Lat cell line and primary CD4+ T cells from both chronic SIV-infected rhesus macaques and HIV-1 patients but without inducing systemic activation, making this compound attractive for potentially treating HIV-1 infection.
Collapse
Affiliation(s)
- Qing Yang
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,State Key Laboratory of Respiratory Disease , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , 510120 , China
| | - Yuyang Ding
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,University of Chinese Academy of Sciences , Beijing , 100049 , China
| | - Fengling Feng
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,School of Life Sciences , University of Science and Technology of China (USTC) , Hefei , 230027 , China
| | - Enxiang Pan
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,University of Chinese Academy of Sciences , Beijing , 100049 , China
| | - Xiaozhen Fan
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,School of Life Sciences , University of Science and Technology of China (USTC) , Hefei , 230027 , China
| | - Xiuchang Ma
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China .
| | - Ling Chen
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China . .,State Key Laboratory of Respiratory Disease , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , 510120 , China
| | - Junling Zhao
- Institute of Medicinal Chemistry , School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P.R. China .
| | - Caijun Sun
- Guangzhou Institutes of Biomedicine and Health (GIBH) , Chinese Academy of Sciences , Guangzhou 510530 , China .
| |
Collapse
|
20
|
Archin NM, Kirchherr JL, Sung JA, Clutton G, Sholtis K, Xu Y, Allard B, Stuelke E, Kashuba AD, Kuruc JD, Eron J, Gay CL, Goonetilleke N, Margolis DM. Interval dosing with the HDAC inhibitor vorinostat effectively reverses HIV latency. J Clin Invest 2017; 127:3126-3135. [PMID: 28714868 DOI: 10.1172/jci92684] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/01/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The histone deacetylase (HDAC) inhibitor vorinostat (VOR) can increase HIV RNA expression in vivo within resting CD4+ T cells of aviremic HIV+ individuals. However, while studies of VOR or other HDAC inhibitors have reported reversal of latency, none has demonstrated clearance of latent infection. We sought to identify the optimal dosing of VOR for effective serial reversal of HIV latency. METHODS In a study of 16 HIV-infected, aviremic individuals, we measured resting CD4+ T cell-associated HIV RNA ex vivo and in vivo following a single exposure to VOR, and then in vivo after a pair of doses separated by 48 or 72 hours, and finally following a series of 10 doses given at 72-hour intervals. RESULTS Serial VOR exposures separated by 72 hours most often resulted in an increase in cell-associated HIV RNA within circulating resting CD4+ T cells. VOR was well tolerated by all participants. However, despite serial reversal of latency over 1 month of VOR dosing, we did not observe a measurable decrease (>0.3 log10) in the frequency of latent infection within resting CD4+ T cells. CONCLUSIONS These findings outline parameters for the experimental use of VOR to clear latent infection. Latency reversal can be achieved by VOR safely and repeatedly, but effective depletion of persistent HIV infection will require additional advances. In addition to improvements in latency reversal, these advances may include the sustained induction of potent antiviral immune responses capable of recognizing and clearing the rare cells in which HIV latency has been reversed. TRIAL REGISTRATION Clinicaltrials.gov NCT01319383. FUNDING NIH grants U01 AI095052, AI50410, and P30 CA016086 and National Center for Advancing Translational Sciences grant KL2 TR001109.
Collapse
Affiliation(s)
- Nancie M Archin
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Jennifer L Kirchherr
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Julia Am Sung
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Genevieve Clutton
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Microbiology and Immunology, UNC Chapel Hill School of Medicine
| | - Katherine Sholtis
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Yinyan Xu
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Brigitte Allard
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Erin Stuelke
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Angela D Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy
| | - Joann D Kuruc
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Joseph Eron
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and.,Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cynthia L Gay
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Nilu Goonetilleke
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Microbiology and Immunology, UNC Chapel Hill School of Medicine
| | - David M Margolis
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and.,Microbiology and Immunology, UNC Chapel Hill School of Medicine.,Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Abstract
OBJECTIVE Resting CD4 T cells have been recognized as the major cell reservoir of latent HIV-1 during antiretroviral therapy (ART). Using an simian immunodeficiency virus (SIV)/macaque model for AIDS and HIV-related neurocognitive disorders we assessed the contribution of the brain to viral latency and reactivation. DESIGN Pigtailed macaques were dual inoculated with SIVDeltaB670 and SIV17E-Fr and treated with an efficacious central nervous system-penetrant ART. After 500 days of viral suppression animals were treated with two cycles of latency reversing agents and increases in viral transcripts were examined. METHODS Longitudinal plasma and cerebrospinal fluid (CSF) viral loads were analyzed by quantitative and digital droplet PCR. After necropsy, viral transcripts in organs were analyzed by PCR, in-situ hybridization, and phylogenetic genotyping based on env V1 loop sequences. Markers for neuronal damage and CSF activation were measured by ELISA. RESULTS Increases in activation markers and plasma and CSF viral loads were observed in one animal treated with latency reversing agents, despite ongoing ART. SIV transcripts were identified in occipital cortex macrophages by in-situ hybridization and CD68 staining. The most abundant SIV genotype in CSF was unique and expanded independent from viruses found in the periphery. CONCLUSION The central nervous system harbors latent SIV genomes after long-term viral suppression by ART, indicating that the brain represents a potential viral reservoir and should be seriously considered during AIDS cure strategies.
Collapse
|
22
|
Manousopoulou A, Saito S, Yamamoto Y, Al-Daghri NM, Ihara M, Carare RO, Garbis SD. Hemisphere Asymmetry of Response to Pharmacologic Treatment in an Alzheimer's Disease Mouse Model. J Alzheimers Dis 2016; 51:333-8. [PMID: 26836196 PMCID: PMC4927832 DOI: 10.3233/jad-151078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of this study was to examine hemisphere asymmetry of response to pharmacologic treatment in an Alzheimer's disease mouse model using cilostazol as a chemical stimulus. Eight-month-old mice were assigned to vehicle or cilostazol treatment for three months and hemispheres were analyzed using quantitative proteomics. Bioinformatics interpretation showed that following treatment, aggregation of blood platelets significantly decreased in the right hemisphere whereas neurodegeneration significantly decreased and synaptic transmission increased in the left hemisphere only. Our study provides novel evidence on cerebral laterality of pharmacologic activity, with important implications in deciphering regional pharmacodynamic effects of existing drugs thus uncovering novel hemisphere-specific therapeutic targets.
Collapse
Affiliation(s)
- Antigoni Manousopoulou
- Institute for Life Sciences, University of Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Satoshi Saito
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Japan
| | - Nasser M Al-Daghri
- Biomarkers Research Program, Department of Biochemistry College of Science, King Saud University, KSA.,Prince Mutaib Chair for Biomarkers of Osteoporosis, Department of Biochemistry King Saud University, KSA
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Japan
| | - Roxana O Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Spiros D Garbis
- Institute for Life Sciences, University of Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK.,Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| |
Collapse
|
23
|
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
|
24
|
Manousopoulou A, Gatherer M, Smith C, Nicoll JAR, Woelk CH, Johnson M, Kalaria R, Attems J, Garbis SD, Carare RO. Systems proteomic analysis reveals that clusterin and tissue inhibitor of metalloproteinases 3 increase in leptomeningeal arteries affected by cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 2016; 43:492-504. [PMID: 27543695 PMCID: PMC5638106 DOI: 10.1111/nan.12342] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 01/06/2023]
Abstract
Aims Amyloid beta (Aβ) accumulation in the walls of leptomeningeal arteries as cerebral amyloid angiopathy (CAA) is a major feature of Alzheimer's disease. In this study, we used global quantitative proteomic analysis to examine the hypothesis that the leptomeningeal arteries derived from patients with CAA have a distinct endophenotypic profile compared to those from young and elderly controls. Methods Freshly dissected leptomeningeal arteries from the Newcastle Brain Tissue Resource and Edinburgh Sudden Death Brain Bank from seven elderly (82.9 ± 7.5 years) females with severe capillary and arterial CAA, as well as seven elderly (88.3 ± 8.6 years) and five young (45.4 ± 3.9 years) females without CAA were used in this study. Arteries from four patients with CAA, two young and two elderly controls were individually analysed using quantitative proteomics. Key proteomic findings were then validated using immunohistochemistry. Results Bioinformatics interpretation of the results showed a significant enrichment of the immune response/classical complement and extracellular matrix remodelling pathways (P < 0.05) in arteries affected by CAA vs. those from young and elderly controls. Clusterin (apolipoprotein J) and tissue inhibitor of metalloproteinases‐3 (TIMP3), validated using immunohistochemistry, were shown to co‐localize with Aβ and to be up‐regulated in leptomeningeal arteries from CAA patients compared to young and elderly controls. Conclusions Global proteomic profiling of brain leptomeningeal arteries revealed that clusterin and TIMP3 increase in leptomeningeal arteries affected by CAA. We propose that clusterin and TIMP3 could facilitate perivascular clearance and may serve as novel candidate therapeutic targets for CAA.
Collapse
Affiliation(s)
- A Manousopoulou
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - M Gatherer
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - C Smith
- Pathology Department, University of Edinburgh, Edinburgh, UK
| | - J A R Nicoll
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - C H Woelk
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - M Johnson
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - R Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - J Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - S D Garbis
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK.,Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - R O Carare
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
25
|
White CH, Moesker B, Ciuffi A, Beliakova-Bethell N. Systems biology applications to study mechanisms of human immunodeficiency virus latency and reactivation. World J Clin Infect Dis 2016; 6:6-21. [DOI: 10.5495/wjcid.v6.i2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/15/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Eradication of human immunodeficiency virus (HIV) in infected individuals is currently not possible because of the presence of the persistent cellular reservoir of latent infection. The identification of HIV latency biomarkers and a better understanding of the molecular mechanisms contributing to regulation of HIV expression might provide essential tools to eliminate these latently infected cells. This review aims at summarizing gene expression profiling and systems biology applications to studies of HIV latency and eradication. Studies comparing gene expression in latently infected and uninfected cells identify candidate latency biomarkers and novel mechanisms of latency control. Studies that profiled gene expression changes induced by existing latency reversing agents (LRAs) highlight uniting themes driving HIV reactivation and novel mechanisms that contribute to regulation of HIV expression by different LRAs. Among the reviewed gene expression studies, the common approaches included identification of differentially expressed genes and gene functional category assessment. Integration of transcriptomic data with other biological data types is presently scarce, and the field would benefit from increased adoption of these methods in future studies. In addition, designing prospective studies that use the same methods of data acquisition and statistical analyses will facilitate a more reliable identification of latency biomarkers using different model systems and the comparison of the effects of different LRAs on host factors with a role in HIV reactivation. The results from such studies would have the potential to significantly impact the process by which candidate drugs are selected and combined for future evaluations and advancement to clinical trials.
Collapse
|
26
|
Hypothalamus proteomics from mouse models with obesity and anorexia reveals therapeutic targets of appetite regulation. Nutr Diabetes 2016; 6:e204. [PMID: 27110685 PMCID: PMC4855256 DOI: 10.1038/nutd.2016.10] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/25/2016] [Accepted: 03/01/2016] [Indexed: 12/27/2022] Open
Abstract
Objective: This study examined the proteomic profile of the hypothalamus in mice exposed to a high-fat diet (HFD) or with the anorexia of acute illness. This comparison could provide insight on the effects of these two opposite states of energy balance on appetite regulation. Methods: Four to six-week-old male C56BL/6J mice were fed a normal (control 1 group; n=7) or a HFD (HFD group; n=10) for 8 weeks. The control 2 (n=7) and lipopolysaccharide (LPS) groups (n=10) were fed a normal diet for 8 weeks before receiving an injection of saline and LPS, respectively. Hypothalamic regions were analysed using a quantitative proteomics method based on a combination of techniques including iTRAQ stable isotope labeling, orthogonal two-dimensional liquid chromatography hyphenated with nanospray ionization and high-resolution mass spectrometry. Key proteins were validated with quantitative PCR. Results: Quantitative proteomics of the hypothalamous regions profiled a total of 9249 protein groups (q<0.05). Of these, 7718 protein groups were profiled with a minimum of two unique peptides for each. Hierachical clustering of the differentiated proteome revealed distinct proteomic signatures for the hypothalamus under the HFD and LPS nutritional conditions. Literature research with in silico bioinformatics interpretation of the differentiated proteome identified key biological relevant proteins and implicated pathways. Furthermore, the study identified potential pharmacologic targets. In the LPS groups, the anorexigen pro-opiomelanocortin was downregulated. In mice with obesity, nuclear factor-κB, glycine receptor subunit alpha-4 (GlyR) and neuropeptide Y levels were elevated, whereas serotonin receptor 1B levels decreased. Conclusions: High-precision quantitative proteomics revealed that under acute systemic inflammation in the hypothalamus as a response to LPS, homeostatic mechanisms mediating loss of appetite take effect. Conversely, under chronic inflammation in the hypothalamus as a response to HFD, mechanisms mediating a sustained ‘perpetual cycle' of appetite enhancement were observed. The GlyR protein may constitute a novel treatment target for the reduction of central orexigenic signals in obesity.
Collapse
|
27
|
Identification of a novel interaction between corticotropin releasing hormone (Crh) and macroautophagy. Sci Rep 2016; 6:23342. [PMID: 26987580 PMCID: PMC4796911 DOI: 10.1038/srep23342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 03/02/2016] [Indexed: 02/07/2023] Open
Abstract
In inflammatory bowel disease (IBD), compromised restitution of the epithelial barrier contributes to disease severity. Owing to the complexity in the pathogenesis of IBD, a variety of factors have been implicated in its progress. In this study, we report a functional interaction between macroautophagy and Corticotropin Releasing Hormone (Crh) in the gut. For this purpose we used DSS colitis model on Crh -/- or wild-type (wt) with pharmacological inhibition of autophagy. We uncovered sustained basal autophagy in the gut of Crh -/- mice, which persisted over the course of DSS administration. Autophagy inhibition resulted in partial rescue of Crh -/- mice, while it increased the expression of Crh in the wt gut. Similarly, Crh deficiency was associated with sustained activation of base line autophagy. In vitro models of amino acid deprivation- and LPS-induced autophagy confirmed the in vivo findings. Our results indicate a novel role for Crh in the intestinal epithelium that involves regulation of autophagy, while suggesting the complementary action of the two pathways. These data suggest the intriguing possibility that targeting Crh stimulation in the intestine may provide a novel therapeutic approach to support the integrity of the epithelial barrier and to protect from chronic colitis.
Collapse
|
28
|
Small Molecule Inhibitors of BAF; A Promising Family of Compounds in HIV-1 Latency Reversal. EBioMedicine 2015; 3:108-121. [PMID: 26870822 PMCID: PMC4739437 DOI: 10.1016/j.ebiom.2015.11.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/19/2015] [Accepted: 11/25/2015] [Indexed: 11/23/2022] Open
Abstract
Persistence of latently infected cells in presence of Anti-Retroviral Therapy presents the main obstacle to HIV-1 eradication. Much effort is thus placed on identification of compounds capable of HIV-1 latency reversal in order to render infected cells susceptible to viral cytopathic effects and immune clearance. We identified the BAF chromatin remodeling complex as a key player required for maintenance of HIV-1 latency, highlighting its potential as a molecular target for inhibition in latency reversal. Here, we screened a recently identified panel of small molecule inhibitors of BAF (BAFi's) for potential to activate latent HIV-1. Latency reversal was strongly induced by BAFi's Caffeic Acid Phenethyl Ester and Pyrimethamine, two molecules previously characterized for clinical application. BAFi's reversed HIV-1 latency in cell line based latency models, in two ex vivo infected primary cell models of latency, as well as in HIV-1 infected patient's CD4 + T cells, without inducing T cell proliferation or activation. BAFi-induced HIV-1 latency reversal was synergistically enhanced upon PKC pathway activation and HDAC-inhibition. Therefore BAFi's constitute a promising family of molecules for inclusion in therapeutic combinatorial HIV-1 latency reversal. BAF complex inhibitors (BAFi's) activate latent HIV-1 in cell line models of latency. BAFi's in combination with HDAC inhibitors and PKC activators synergistically activate latent HIV-1. The BAFi's PYR and CAPE reverse HIV-1 latency in primary cell models of latency and in cells obtained from HIV-1 patients.
Access to Combination antiretroviral therapy (cART) has made HIV-1 infection a chronic disease. However, cART is not curative, as a small number of infected cells harboring silent virus with potential to renew the infection persist despite cART. Efforts to cure HIV-1 include activation of these latent cells, making them susceptible to immune clearance. Here we describe the activity of BAF inhibitors in HIV-1 activation in in vitro models of HIV-1 latency as well as in cells obtained from HIV-1 infected patient volunteers. Our data highlight the clinical potential of BAF inhibitors for inclusion in combinatorial therapy to reverse HIV-1 latency.
Collapse
Key Words
- BAF complex
- BAF, BRG-Brahma Associated Factors
- BAF250a, BAF Associated Factor 250 a
- BAFi, BAF inhibitor
- BRG-1, Brahma Related Gene 1
- CAPE, caffeic acid phenetyl esther
- ChIP, Chromatin Immunoprecipitation
- Chromatin remodeling
- CycA, Cyclophilin A
- DHS-1, DNase Hypersensitive Site 1
- ES cells, embryonic stem cells
- FAIRE, Formaldehyde Assisted Isolation of Regulatory Elements
- FBS, Fetal Bovine Serum
- GFP, Green Fluorescent Protein
- HDAC, histone deacetylase
- HIV
- HIV-1, human immunodeficiency virus type 1
- IFNß, Interferon beta
- IL10, Interleukin 10
- IL12, Interleukin 12
- IL4, Interleukin 4
- IL6, Interleukin 6
- INI-1, Integrase Interactor 1
- IRES, Internal Ribosome Entry Site
- IκB-α, Inhibitor of NF-κB – alpha
- LRA, latency reversal agent
- LTR, Long Terminal Repeat
- Latency
- MIP26, Major Intrinsic Protein
- MMP9, Matrix Metallopeptidase 9
- NF-κB, Nuclear Factor Kappa-light-chain-enhancer of activated B cells
- PBMC, peripheral blood mononuclear cell
- PBS, Phosphate Buffered Saline
- PKC, Protein Kinase C
- PYR, Pyrimethamine
- RT-qPCR, Reverse Transcriptase, quantitative Polymerase Chain Reaction
- SAHA, Suberoylanilide Hydroxamic Acid
- SD, Standard Deviation
- SOCS3, Suppressor Of Cytokine Signaling 3
- TGF-ß, Transforming Growth Factor beta
- TLR2, Toll-like Receptor 2
- bp, base pairs
- cART, combination Antiretroviral Therapy
- latency reversal agents
- nuc, nucleosome
- siRNA, small interfering RNA
Collapse
|
29
|
Zhang Y, Yin C, Zhang T, Li F, Yang W, Kaminski R, Fagan PR, Putatunda R, Young WB, Khalili K, Hu W. CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs. Sci Rep 2015; 5:16277. [PMID: 26538064 PMCID: PMC4633726 DOI: 10.1038/srep16277] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 01/04/2023] Open
Abstract
Current antiretroviral therapy does not eliminate the integrated and transcriptionally silent HIV-1 provirus in latently infected cells. Recently, a "shock and kill" strategy has been extensively explored to eradicate the HIV-1 latent reservoirs for a permanent cure of AIDS. The therapeutic efficacy of currently used agents remains disappointing because of low efficiency, non-specificity and cellular toxicity. Here we present a novel catalytically-deficient Cas9-synergistic activation mediator (dCas9-SAM) technology to selectively, potently and persistently reactivate the HIV-1 latent reservoirs. By screening 16 MS2-mediated single guide RNAs, we identified long terminal repeat (LTR)-L and O that surround the enhancer region (-165/-145 for L and -92/-112 for O) and induce robust reactivation of HIV-1 provirus in HIV-1 latent TZM-bI epithelial, Jurkat T lymphocytic and CHME5 microglial cells. This compulsory reactivation induced cellular suicide via toxic buildup of viral proteins within HIV-1 latent Jurkat T and CHME5 microglial cells. These results suggest that this highly effective and target-specific dCas9-SAM system can serve as a novel HIV-latency-reversing therapeutic tool for the permanent elimination of HIV-1 latent reservoirs.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Chaoran Yin
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Ting Zhang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Fang Li
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Wensheng Yang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Rafal Kaminski
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Philip Regis Fagan
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Raj Putatunda
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Won-Bin Young
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Wenhui Hu
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| |
Collapse
|
30
|
Modeling the Effects of Vorinostat In Vivo Reveals both Transient and Delayed HIV Transcriptional Activation and Minimal Killing of Latently Infected Cells. PLoS Pathog 2015; 11:e1005237. [PMID: 26496627 PMCID: PMC4619772 DOI: 10.1371/journal.ppat.1005237] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/28/2015] [Indexed: 12/31/2022] Open
Abstract
Recent efforts to cure human immunodeficiency virus type-1 (HIV-1) infection have focused on developing latency reversing agents as a first step to eradicate the latent reservoir. The histone deacetylase inhibitor, vorinostat, has been shown to activate HIV RNA transcription in CD4+ T-cells and alter host cell gene transcription in HIV-infected individuals on antiretroviral therapy. In order to understand how latently infected cells respond dynamically to vorinostat treatment and determine the impact of vorinostat on reservoir size in vivo, we have constructed viral dynamic models of latency that incorporate vorinostat treatment. We fitted these models to data collected from a recent clinical trial in which vorinostat was administered daily for 14 days to HIV-infected individuals on suppressive ART. The results show that HIV transcription is increased transiently during the first few hours or days of treatment and that there is a delay before a sustained increase of HIV transcription, whose duration varies among study participants and may depend on the long term impact of vorinostat on host gene expression. Parameter estimation suggests that in latently infected cells, HIV transcription induced by vorinostat occurs at lower levels than in productively infected cells. Furthermore, the estimated loss rate of transcriptionally induced cells remains close to baseline in most study participants, suggesting vorinostat treatment does not induce latently infected cell killing and thus reduce the latent reservoir in vivo. Combination antiretroviral therapy (cART) for HIV infection must be taken for life due to the existence of long lived latently infected cells. Recent efforts have focused on developing latency reversing agents to eliminate latently infected cells by activating HIV production. In this work, we assess the impact of a latency reversing agent, vorinostat, by fitting dynamic models to data from a clinical trial. Results show that vorinostat treatment induces HIV transcription transiently and that the sustained induction of HIV transcription may depend on the temporal impact of vorinostat on host gene expression. Our results also suggest that vorinostat treatment is not sufficient to induce killing of latently infected cells in a majority of HIV-infected individuals on cART.
Collapse
|