1
|
Zenere G, Wu C, Midkiff CC, Johnson NM, Grice CP, Wimley WC, Kaur A, Braun SE. Extracellular domain, hinge, and transmembrane determinants affecting surface CD4 expression of a novel anti-HIV chimeric antigen receptor (CAR) construct. PLoS One 2024; 19:e0293990. [PMID: 39133676 PMCID: PMC11318886 DOI: 10.1371/journal.pone.0293990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated clinical potential, but current receptors still need improvements to be successful against chronic HIV infection. In this study, we address some requirements of CAR motifs for strong surface expression of a novel anti-HIV CAR by evaluating important elements in the extracellular, hinge, and transmembrane (TM) domains. When combining a truncated CD4 extracellular domain and CD8α hinge/TM, the novel CAR did not express extracellularly but was detectable intracellularly. By shortening the CD8α hinge, CD4-CAR surface expression was partially recovered and addition of the LYC motif at the end of the CD8α TM fully recovered both intracellular and extracellular CAR expression. Mutation of LYC to TTA or TTC showed severe abrogation of CAR expression by flow cytometry and confocal microscopy. Additionally, we determined that CD4-CAR surface expression could be maximized by the removal of FQKAS motif at the junction of the extracellular domain and the hinge region. CD4-CAR surface expression also resulted in cytotoxic CAR T cell killing of HIV Env+ target cells. In this study, we identified elements that are crucial for optimal CAR surface expression, highlighting the need for structural analysis studies to establish fundamental guidelines of CAR designs.
Collapse
Affiliation(s)
- Giorgio Zenere
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Chengxiang Wu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Cecily C. Midkiff
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Nathan M. Johnson
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Christopher P. Grice
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - William C. Wimley
- Department of BioChemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Stephen E. Braun
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
2
|
Zenere G, Wu C, Midkiff CC, Johnson NM, Grice CP, Wimley WC, Kaur A, Braun SE. Extracellular domain, hinge, and transmembrane determinants affecting surface CD4 expression of a novel anti-HIV chimeric antigen receptor (CAR) construct. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.25.563930. [PMID: 37961145 PMCID: PMC10634810 DOI: 10.1101/2023.10.25.563930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated clinical potential, but current receptors still need improvements to be successful against chronic HIV infection. In this study, we address some requirements of CAR motifs for strong surface expression of a novel anti-HIV CAR by evaluating important elements in the extracellular, hinge, and transmembrane (TM) domains. When combining a truncated CD4 extracellular domain and CD8α hinge/TM, the novel CAR did not express extracellularly but was detectable intracellularly. By shortening the CD8α hinge, CD4-CAR surface expression was partially recovered and addition of the LYC motif at the end of the CD8α TM fully recovered both intracellular and extracellular CAR expression. Mutation of LYC to TTA or TTC showed severe abrogation of CAR expression by flow cytometry and confocal microscopy. Additionally, we determined that CD4-CAR surface expression could be maximized by the removal of FQKAS motif at the junction of the extracellular domain and the hinge region. CD4-CAR surface expression also resulted in cytotoxic CAR T cell killing of HIV Env+ target cells. In this study, we identified elements that are crucial for optimal CAR surface expression, highlighting the need for structural analysis studies to establish fundamental guidelines of CAR designs.
Collapse
Affiliation(s)
- Giorgio Zenere
- Tulane National Primate Research Center, Covington, LA 70433
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, LA 70112
| | - Chengxiang Wu
- Tulane National Primate Research Center, Covington, LA 70433
| | | | - Nathan M. Johnson
- Tulane National Primate Research Center, Covington, LA 70433
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, LA 70112
| | - Christopher P. Grice
- Tulane National Primate Research Center, Covington, LA 70433
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112
| | - William C. Wimley
- Department of BioChemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Amitinder Kaur
- Tulane National Primate Research Center, Covington, LA 70433
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Stephen E. Braun
- Tulane National Primate Research Center, Covington, LA 70433
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112
| |
Collapse
|
3
|
Eldesouki RE, Wu C, Saleh FM, Mohammed EAM, Younes S, Hassan NE, Brown TC, Alt EU, Robinson JE, Badr FM, Braun SE. Identification and Targeting of Thomsen-Friedenreich and IL1RAP Antigens on Chronic Myeloid Leukemia Stem Cells Using Bi-Specific Antibodies. Onco Targets Ther 2021; 14:609-621. [PMID: 33519209 PMCID: PMC7837560 DOI: 10.2147/ott.s255299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Quiescent leukemia stem cells (LSCs) play a major role in therapeutic resistance and disease progression of chronic myeloid leukemia (CML). LSCs belong to the primitive population; CD34+CD38-Lin-, which does not distinguish normal hematopoietic stem cells (HSC) from CML LSCs. Because Thomsen–Friedenreich/CD176 antigen is expressed on CD34+ HSC and IL1RAP is tightly correlated to BCR-ABL expression, we sought to increase the specificity towards LSC by using additional biomarkers. Methods We evaluated the co-expression of both antigens on CD34+ peripheral blood mononuclear cells (PBMCs) from both healthy volunteers and CML patients, using flow cytometry. Then, we used site-directed mutagenesis to induce knob-in-hole mutations in the human IgG heavy chain and the human lambda light chain to generate the bi-specific antibody (Bis-Ab) TF/RAP that binds both antigens simultaneously. We measured complement-directed cytotoxicity (CDC) in CML samples with the Bis-Ab by flow cytometry. Results In contrast to healthy volunteers, CML samples displayed a highly significant co-expression of CD176 and IL1RAP. When either a double-positive cell line or CML samples were treated with increasing doses of Bis-Ab, increased binding and CDC was observed indicating co-operative binding of the Bis-Ab as compared to monoclonal antibodies. Discussion These results show that the bi-specific antibody is capable of targeting IL1RAP+ and CD176+ cell population among CML PBMCs, but not corresponding normal cells in CDC assay. We hereby offer a novel strategy for the depletion of CML stem cells from the bulk population in clinical hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Raghda E Eldesouki
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt.,Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| | - Chengxiang Wu
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| | - Fayez M Saleh
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA.,Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Eman Abdel-Moemen Mohammed
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt
| | - Soha Younes
- Department of Clinical pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Theresa C Brown
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Eckhard U Alt
- Applied Stem Cell Laboratory, Departments of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - James E Robinson
- Sections of Infectious Disease, Departments of Pediatrics and Internal Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Fouad Mohamed Badr
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt
| | - Stephen E Braun
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA.,Departments of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
4
|
Chen K, Wang S, Sun QW, Zhang B, Ullah M, Sun Z. Klotho Deficiency Causes Heart Aging via Impairing the Nrf2-GR Pathway. Circ Res 2020; 128:492-507. [PMID: 33334122 DOI: 10.1161/circresaha.120.317348] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Cardiac aging is an important contributing factor for heart failure, which affects a large population but remains poorly understood. OBJECTIVE The purpose of this study is to investigate whether Klotho plays a role in cardiac aging. METHODS AND RESULTS Heart function declined in old mice (24 months), as evidenced by decreases in fractional shortening, ejection fraction, and cardiac output. Heart size and weight, cardiomyocyte size, and cardiac fibrosis were increased in old mice, indicating that aging causes cardiac hypertrophy and remodeling. Circulating Klotho levels were dramatically decreased in old mice, which prompted us to investigate whether the Klotho decline may cause heart aging. We found that Klotho gene mutation (KL-/-) largely decreased serum klotho levels and impaired heart function. Interestingly, supplement of exogenous secreted Klotho prevented heart failure, hypertrophy, and remodeling in both old mice and KL (-/-) mice. Secreted Klotho treatment inhibited excessive cardiac oxidative stress, senescence and apoptosis in old mice and KL (-/-) mice. Serum phosphate levels in KL (-/-) mice were kept in the normal range, suggesting that Klotho deficiency-induced heart aging is independent of phosphate metabolism. Mechanistically, Klotho deficiency suppressed GR (glutathione reductase) expression and activity in the heart via inhibition of transcription factor Nrf2 (nuclear factor-erythroid 2 p45-related factor 2). Furthermore, cardiac-specific overexpression of GR prevented excessive oxidative stress, apoptosis, and heart failure in both old and KL (-/-) mice. CONCLUSIONS Klotho deficiency causes cardiac aging via impairing the Nrf2-GR pathway. Supplement of exogenous secreted Klotho represents a promising therapeutic strategy for aging-associated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., S.W., Q.W.S., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (K.C., S.W., M.U., Z.S.)
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., S.W., Q.W.S., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (K.C., S.W., M.U., Z.S.)
| | - Qiwei Wilton Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., S.W., Q.W.S., B.Z., Z.S.)
| | - Bo Zhang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., S.W., Q.W.S., B.Z., Z.S.)
| | - Mujib Ullah
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (K.C., S.W., M.U., Z.S.)
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., S.W., Q.W.S., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (K.C., S.W., M.U., Z.S.)
| |
Collapse
|
5
|
Ghani K, Boivin-Welch M, Roy S, Dakiw-Piaceski A, Barbier M, Pope E, Germain L, Caruso M. Generation of High-Titer Self-Inactivated γ-Retroviral Vector Producer Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:90-99. [PMID: 31312667 PMCID: PMC6610700 DOI: 10.1016/j.omtm.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/30/2019] [Indexed: 12/01/2022]
Abstract
The γ-retroviral vector is a gene delivery vehicle that is commonly used in gene therapy. Despite its efficacy, its strong enhancers contributed to malignant transformations in some hematopoietic stem cell (HSC) gene therapy trials. A safer version without viral enhancers (SIN) is available, but its production is cumbersome, as high titers can only be obtained in transient transfection. Our aim was to develop a system that could easily generate high-titer SIN vectors from stable producer cells. The use of the cytomegalovirus enhancer-promoter sequence to generate the full-length genomic RNA combined to sequences that decrease transcriptional readthrough (WPRE and strong polyadenylation sequences) led to 6 × 106 infectious units (IU)/mL of a SIN GFP vector in transient transfection. The incorporation of a blasticidin selection cassette to the retroviral plasmid allowed the generation of stable clones in the 293Vec packaging cells that release 2 × 107 IU/mL and 1.4 × 107 IU/mL of a SIN GFP and a SIN PIGA vector, respectively. A titer of 1.8 × 106 IU/mL was obtained with a SIN vector containing the long 8.9-kb COL7A1 cDNA. Thus, an efficient process was established for the generation of stable 293Vec-derived retrovirus producer cells that release high-titer SIN vectors.
Collapse
Affiliation(s)
- Karim Ghani
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Michael Boivin-Welch
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada.,CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Sylvie Roy
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Angela Dakiw-Piaceski
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Martin Barbier
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Elena Pope
- Section of Dermatology, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Lucie Germain
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| |
Collapse
|
6
|
Wang B, Zuo J, Kang W, Wei Q, Li J, Wang C, Liu Z, Lu Y, Zhuang Y, Dang B, Liu Q, Kang W, Sun Y. Generation of Hutat2:Fc Knockin Primary Human Monocytes Using CRISPR/Cas9. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:130-141. [PMID: 29858049 PMCID: PMC5992333 DOI: 10.1016/j.omtn.2018.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 10/28/2022]
Abstract
The ability of monocytes to travel through the bloodstream, traverse tissue barriers, and aggregate at disease sites endows these cells with the attractive potential to carry therapeutic genes into the nervous system. However, gene editing in primary human monocytes has long been a challenge. Here, we applied the CRISPR/Cas9 system to deliver the large functional Hutat2:Fc DNA fragment into the genome of primary monocytes to neutralize HIV-1 transactivator of transcription (Tat), an essential neurotoxic factor that causes HIV-associated neurocognitive disorder (HAND) in the nervous system. Following homology-directed repair (HDR), ∼10% of the primary human monocytes exhibited knockin of the Hutat2:Fc gene in the AAVS1 locus, the "safe harbor" locus of the human genome, without selection. Importantly, the release of Hutat2:Fc by these modified monocytes protected neurons from Tat-induced neurotoxicity, reduced HIV replication, and restored T cell homeostasis. Moreover, compared with lentiviral transfection, CRISPR-mediated knockin had the advantage of maintaining the migrating function of monocytes. These results establish CRISPR/Cas9-mediated Hutat2:Fc knockin monocytes and provide a potential method to cross the blood-brain barrier for HAND therapy.
Collapse
Affiliation(s)
- Bowen Wang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Jiahui Zuo
- Clinical Laboratory, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Wenzhen Kang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Qianqi Wei
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Jianhui Li
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Chunfu Wang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Zhihui Liu
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Yuanan Lu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI 96822, USA
| | - Yan Zhuang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Bianli Dang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Qing Liu
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Wen Kang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China; Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI 96822, USA.
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
7
|
Lee H, Radu C, Han JW, Grailhe R. Assay Development for High Content Quantification of Sod1 Mutant Protein Aggregate Formation in Living Cells. J Vis Exp 2017. [PMID: 29053667 DOI: 10.3791/56425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that can be caused by inherited mutations in the gene encoding copper-zinc superoxide dismutase 1 (SOD1). The structural instability of SOD1 and the detection of SOD1-positive inclusions in familial-ALS patients supports a potential causal role for misfolded and/or aggregated SOD1 in ALS pathology. In this study, we describe the development of a cell-based assay designed to quantify the dynamics of SOD1 aggregation in living cells by high content screening approaches. Using lentiviral vectors, we generated stable cell lines expressing wild-type and mutant A4V SOD1 tagged with yellow fluorescent protein and found that both proteins were expressed in the cytosol without any sign of aggregation. Interestingly, only SOD1 A4V stably expressed in HEK-293, but not in U2OS or SH-SY5Y cell lines, formed aggregates upon proteasome inhibitor treatment. We show that it is possible to quantify aggregation based on dose-response analysis of various proteasome inhibitors, and to track aggregate-formation kinetics by time-lapse microscopy. Our approach introduces the possibility of quantifying the effect of ALS mutations on the role of SOD1 in aggregate formation as well as screening for small molecules that prevent SOD1 A4V aggregation.
Collapse
Affiliation(s)
- Honggun Lee
- Automation & Logistics Management, Screening Sciences & Novel Assay Technologies, Institut Pasteur Korea
| | - Constantin Radu
- Automation & Logistics Management, Screening Sciences & Novel Assay Technologies, Institut Pasteur Korea
| | | | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea;
| |
Collapse
|
8
|
Kaljas Y, Liu C, Skaldin M, Wu C, Zhou Q, Lu Y, Aksentijevich I, Zavialov AV. Human adenosine deaminases ADA1 and ADA2 bind to different subsets of immune cells. Cell Mol Life Sci 2017; 74:555-570. [PMID: 27663683 PMCID: PMC11107696 DOI: 10.1007/s00018-016-2357-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 11/29/2022]
Abstract
At sites of inflammation and tumor growth, the local concentration of extracellular adenosine rapidly increases and plays a role in controlling the immune responses of nearby cells. Adenosine deaminases ADA1 and ADA2 (ADAs) decrease the level of adenosine by converting it to inosine, which serves as a negative feedback mechanism. Mutations in the genes encoding ADAs lead to impaired immune function, which suggests a crucial role for ADAs in immune system regulation. It is not clear why humans and other mammals possess two enzymes with adenosine deaminase activity. Here, we found that ADA2 binds to neutrophils, monocytes, NK cells and B cells that do not express CD26, a receptor for ADA1. Moreover, the analysis of CD4+ T-cell subset revealed that ADA2 specifically binds to regulatory T cells expressing CD39 and lacking the receptor for ADA1. Also, it was found that ADA1 binds to CD16- monocytes, while CD16+ monocytes preferably bind ADA2. A study of the blood samples from ADA2-deficient patients showed a dramatic reduction in the number of lymphocyte subsets and an increased concentration of TNF-α in plasma. Our results suggest the existence of a new mechanism, where the activation and survival of immune cells is regulated through the activities of ADA2 or ADA1 anchored to the cell surface.
Collapse
Affiliation(s)
- Yuliia Kaljas
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, 9 Jinsui Road, Tianhe, Guangzhou, 510623, Guangdong, China
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520, Turku, Finland
| | - Chengqian Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, 9 Jinsui Road, Tianhe, Guangzhou, 510623, Guangdong, China
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520, Turku, Finland
| | - Maksym Skaldin
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, 9 Jinsui Road, Tianhe, Guangzhou, 510623, Guangdong, China
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520, Turku, Finland
| | - Chengxiang Wu
- Department of Public Health, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East West Road, Biomed, Honolulu, HI, 96822, USA
- Division of Regenerative Medicine, Tulane National Primate Research Center, 18703 Three River Road, Covington, LA, 70433, USA
| | - Qing Zhou
- National Institutes of Health, National Human Genome Research Institute, Bldg. 10, Bethesda, MD, 20892, USA
| | - Yuanan Lu
- Department of Public Health, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East West Road, Biomed, Honolulu, HI, 96822, USA
| | - Ivona Aksentijevich
- Department of Public Health, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East West Road, Biomed, Honolulu, HI, 96822, USA
| | - Andrey V Zavialov
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, 9 Jinsui Road, Tianhe, Guangzhou, 510623, Guangdong, China.
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520, Turku, Finland.
| |
Collapse
|
9
|
Liu C, Skaldin M, Wu C, Lu Y, Zavialov AV. Application of ADA1 as a new marker enzyme in sandwich ELISA to study the effect of adenosine on activated monocytes. Sci Rep 2016; 6:31370. [PMID: 27510152 PMCID: PMC4980770 DOI: 10.1038/srep31370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/18/2016] [Indexed: 01/24/2023] Open
Abstract
Enzyme-linked immunosorbent assay (ELISA) is a valuable technique to detect antigens in biological fluids. Horse radish peroxidase (HRP) is one of the most common enzymes used for signal amplification in ELISA. Despite new advances in technology, such as a large-scale production of recombinant enzymes and availability of new detection systems, limited research is devoted to finding alternative enzymes and their substrates to amplify the ELISA signals. Here, HRP-avidin was substituted with the human adenosine deaminase (hADA1)-streptavidin complex and adenosine as a detection system in commercial ELISA kits. The hADA1 ELISA was successfully used to demonstrate that adenosine, bound to A1 and A3 adenosine receptors, increases cytokine secretion by LPS activated monocytes. We show that hADA1-based ELISA has the same sensitivity, and also provides identical results, as HRP ELISA. In addition, the sensitivity of hADA1-based ELISA could be easily adjusted by changing the adenosine concentration and the incubation time. Therefore, hADA1 could be used as a detection enzyme with any commercial ELISA kit with a wide range of concentration of antigens.
Collapse
Affiliation(s)
- Chengqian Liu
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520 Turku, Finland
| | - Maksym Skaldin
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520 Turku, Finland
| | - Chengxiang Wu
- Tulane National Primate Research Center, 18703 Three River Road, Covington, LA 70433, USA.,Department of Public Health, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East West Road, Biomed, Honolulu, HI 96822, USA
| | - Yuanan Lu
- Department of Public Health, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East West Road, Biomed, Honolulu, HI 96822, USA
| | - Andrey V Zavialov
- Turku Centre for Biotechnology, University of Turku, Tykistokatu 6, 20520 Turku, Finland
| |
Collapse
|
10
|
Shi Y, Xiang J, Zhou G, Ron TB, Tong HI, Kang W, Sun S, Lu Y. The Pacific White Shrimp β-actin Promoter: Functional Properties and the Potential Application for Transduction System Using Recombinant Baculovirus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2016; 18:349-358. [PMID: 27177910 DOI: 10.1007/s10126-016-9700-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/28/2016] [Indexed: 06/05/2023]
Abstract
A newly isolated Pacific white shrimp (Litopenaeus vannamei) beta-actin promoter SbaP and its derivative compact construct SbaP (ENX) have recently been demonstrated to promote ectopic gene expression in vitro and in vivo. To further explore the potential transduction application, this newly isolated shrimp promoter SbaP was comparatively tested with cytomegalovirus (CMV), simian virus 40 (SV40), polyhedrin (Polh), and white spot syndrome virus immediate early gene 1 (WSSV ie1) four constitutive promoters and a beta-actin promoter (TbaP) from tilapia fish to characterize its promoting function in eight different cell lines. Luciferase quantitation assays revealed that SbaP can drive luciferase gene expression in all eight cell lines including sf21 (insect), PAC2 (zebrafish), EPC (carp), CHSE-214 (chinook salmon), GSTEF (green sea turtle), MS-1 (monk seal), 293T (human), and HeLa (human), but at different levels. Comparative analysis revealed that the promoting activity of SbaP was lower (≤10-fold) than CMV but higher (2-20 folds) than Polh in most of these cell lines tested. Whereas, SbaP mediated luciferase expression in sf21 cells was over 20-fold higher than CMV, SV40, Polh, and TbaP promoter. Compared to the SbaP, SbaP (ENX), which was constructed on the basis of SbaP by deletion of two "negative" regulatory elements, exhibited no significant change of promoting activity in EPC and PAC2 cells, but a 5 and 16 % lower promoting effect in 293T and HeLa cells, respectively. Additionally, a recombinant baculovirus was constructed under the control of SbaP (ENX), and efficient promoter activity of newly generated baculoviral vector was detected both in vitro of infected sf21 cells and in vivo of injected indicator shrimp. These results warrant the potential application of SbaP, particularly SbaP (ENX) in ectopic gene expression in future.
Collapse
Affiliation(s)
- Yingli Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Jianhai Xiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.
| | - Guangzhou Zhou
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, China
| | - Tetsuzan Benny Ron
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Hsin-I Tong
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Wen Kang
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Si Sun
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Yuanan Lu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
11
|
Kang W, Marasco WA, Tong HI, Byron MM, Wu C, Shi Y, Sun S, Sun Y, Lu Y. Anti-tat Hutat2:Fc mediated protection against tat-induced neurotoxicity and HIV-1 replication in human monocyte-derived macrophages. J Neuroinflammation 2014; 11:195. [PMID: 25416164 PMCID: PMC4256057 DOI: 10.1186/s12974-014-0195-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/05/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND HIV-1 Tat is essential for HIV replication and is also a well-known neurotoxic factor causing HIV-associated neurocognitive disorder (HAND). Currently, combined antiretroviral therapy targeting HIV reverse transcriptase or protease cannot prevent the production of early viral proteins, especially Tat, once HIV infection has been established. HIV-infected macrophages and glial cells in the brain still release Tat into the extracellular space where it can exert direct and indirect neurotoxicity. Therefore, stable production of anti-Tat antibodies in the brain would neutralize HIV-1 Tat and thus provide an effective approach to protect neurons. METHODS We constructed a humanized anti-Tat Hutat2:Fc fusion protein with the goal of antagonizing HIV-1 Tat and delivered the gene into cell lines and primary human monocyte-derived macrophages (hMDM) by an HIV-based lentiviral vector. The function of the anti-Tat Hutat2:Fc fusion protein and the potential side effects of lentiviral vector-mediated gene transfer were evaluated in vitro. RESULTS Our study demonstrated that HIV-1-based lentiviral vector-mediated gene transduction resulted in a high-level, stable expression of anti-HIV-1 Tat Hutat2:Fc in human neuronal and monocytic cell lines, as well as in primary hMDM. Hutat2:Fc was detectable in both cells and supernatants and continued to accumulate to high levels within the supernatant. Hutat2:Fc protected mouse cortical neurons against HIV-1 Tat86-induced neurotoxicity. In addition, both secreted Hutat2:Fc and transduced hMDM led to reducing HIV-1BaL viral replication in human macrophages. Moreover, lentiviral vector-based gene introduction did not result in any significant changes in cytomorphology and cell viability. Although the expression of IL8, STAT1, and IDO1 genes was up-regulated in transduced hMDM, such alternation in gene expression did not affect the neuroprotective effect of Hutat2:Fc. CONCLUSIONS Our study demonstrated that lentivirus-mediated gene transfer could efficiently deliver the Hutat2:Fc gene into primary hMDM and does not lead to any significant changes in hMDM immune-activation. The neuroprotective and HIV-1 suppressive effects produced by Hutat2:Fc were comparable to that of a full-length anti-Tat antibody. This study provides the foundation and insights for future research on the potential use of Hutat2:Fc as a novel gene therapy approach for HAND through utilizing monocytes/macrophages, which naturally cross the blood-brain barrier, for gene delivery.
Collapse
Affiliation(s)
- Wen Kang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China. .,Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 50 Brookline Avenue, Boston, MA, 02215, USA.
| | - Hsin-I Tong
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Mary Margaret Byron
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St., BSB, Suite 231, Honolulu, HI, 96813, USA.
| | - Chengxiang Wu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yingli Shi
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Si Sun
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China.
| | - Yuanan Lu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| |
Collapse
|
12
|
Tong J, Buch S, Yao H, Wu C, Tong HI, Wang Y, Lu Y. Monocytes-derived macrophages mediated stable expression of human brain-derived neurotrophic factor, a novel therapeutic strategy for neuroAIDS. PLoS One 2014; 9:e82030. [PMID: 24505242 PMCID: PMC3914783 DOI: 10.1371/journal.pone.0082030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 10/19/2013] [Indexed: 12/30/2022] Open
Abstract
HIV-1 associated dementia remains a significant public health burden. Clinical and experimental research has shown that reduced levels of brain-derived neurotrophic factor (BDNF) may be a risk factor for neurological complications associated with HIV-1 infection. We are actively testing genetically modified macrophages for their possible use as the cell-based gene delivery vehicle for the central nervous system (CNS). It can be an advantage to use the natural homing/migratory properties of monocyte-derived macrophages to deliver potentially neuroprotective BDNF into the CNS, as a non-invasive manner. Lentiviral-mediated gene transfer of human (h)BDNF plasmid was constructed and characterized. Defective lentiviral stocks were generated by transient transfection of 293T cells with lentiviral transfer plasmid together with packaging and envelope plasmids. High titer lentiviral vector stocks were harvested and used to transduce human neuronal cell lines, primary cultures of human peripheral mononocyte-derived macrophages (hMDM) and murine myeloid monocyte-derived macrophages (mMDM). These transduced cells were tested for hBDNF expression, stability, and neuroprotective activity. The GenomeLab GeXP Genetic Analysis System was used to evaluate transduced cells for any adverse effects by assessing gene profiles of 24 reference genes. High titer vectors were prepared for efficient transduction of neuronal cell lines, hMDM, and mMDM. Stable secretion of high levels of hBDNF was detected in supernatants of transduced cells using western blot and ELISA. The conditioned media containing hBDNF were shown to be protective to neuronal and monocytic cell lines from TNF-α and HIV-1 Tat mediated cytotoxicity. Lentiviral vector-mediated gene transduction of hMDM and mMDM resulted in high-level, stable expression of the neuroprotective factorBDNF in vitro. These findings form the basis for future research on the potential use of BDNF as a novel therapy for neuroAIDS.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan, People's Republic of China
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Shilpa Buch
- University of Nebraska Medical Center, Pharmacology and Experimental Neuroscience, Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Honghong Yao
- University of Nebraska Medical Center, Pharmacology and Experimental Neuroscience, Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chengxiang Wu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Hsin-I Tong
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Youwei Wang
- MOE Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan, People's Republic of China
- * E-mail: (YW); (YL)
| | - Yuanan Lu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- * E-mail: (YW); (YL)
| |
Collapse
|
13
|
Wu C, Nerurkar VR, Lu Y. New insights into inhibition of human immunodeficiency virus type 1 replication through mutant tRNALys3. Retrovirology 2013; 10:112. [PMID: 24156557 PMCID: PMC3832253 DOI: 10.1186/1742-4690-10-112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 09/25/2013] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Host cellular tRNA(Lys3) is exclusively utilized by human immunodeficiency virus type 1 (HIV-1) as a primer for the replication step of reverse transcription (RTion). Consequently, the priming step of HIV-1 RT constitutes a potential target for anti-HIV-1 intervention. Previous studies indicated that a mutant tRNA(Lys3) with 7-nucleotide substitutions in the 3' terminus resulted in aberrant HIV-1 RTion from the trans-activation response region (TAR) and inhibition of HIV-1 replication. However, the mutant tRNA(Lys3) also directed HIV-1 RTion from the normal primer-binding site (PBS) with potentially weakened anti-HIV-1 activity. To achieve improved targeting of HIV-1 RTion at sites not including the PBS, a series of mutant tRNA(Lys3) with extended lengths of mutations containing up to 18 bases complementary to their targeting sites were constructed and characterized. RESULTS A positive correlation between the length of mutation in the 3' PBS-binding region of tRNA(Lys3) and the specificity of HIV-1 RTion initiation from the targeting site was demonstrated, as indicated by the potency of HIV-1 inhibition and results of priming assays. Moreover, two mutant tRNA(Lys3)s that targeted the IN-encoding region and Env gene, respectively, both showed a high anti-HIV-1 activity, suggesting that not only the TAR, but also distant sites downstream of the PBS could be effectively targeted by mutant tRNA(Lys3). To increase the expression of mutant tRNA(Lys3), multiple-copy expression cassettes were introduced into target cells with increased anti-HIV-1 potency. CONCLUSIONS These results highlight the importance of the length of complementarity between the 3' terminus of the mutant tRNA(Lys3) and its target site, and the feasibility of targeting multiple sites within the HIV-1 genome through mutant tRNA(Lys3). Intervention of the HIV-1 genome conversion through mutant tRNA(Lys3) may constitute an effective approach for development of novel therapeutics against HIV-1 replication and HIV-1-associated diseases.
Collapse
Affiliation(s)
- Chengxiang Wu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East–west Road, Biomed. Bldg, D105, Honolulu, Hawaii 96822, USA
- Department of Microbiology, College of Natural Sciences, University of Hawaii at Manoa, 2538 McCarthy Mall, Snyder 207, Honolulu, HI 96822, USA
| | - Vivek R Nerurkar
- Departments of Tropical Medicine, Medical Microbiology and Pharmacology, Asia-Pacific Institute of Tropical Medicine and Infectious Diseases. John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, BSB 325AA, Honolulu, HI 96813, USA
| | - Yuanan Lu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, 1960 East–west Road, Biomed. Bldg, D105, Honolulu, Hawaii 96822, USA
| |
Collapse
|
14
|
Qin Q, Lee SH, Liang R, Kalejta RF. Insertion of myeloid-active elements into the human cytomegalovirus major immediate early promoter is not sufficient to drive its activation upon infection of undifferentiated myeloid cells. Virology 2013; 448:125-32. [PMID: 24314643 DOI: 10.1016/j.virol.2013.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 07/29/2013] [Accepted: 10/07/2013] [Indexed: 12/13/2022]
Abstract
The Major Immediate Early Promoter (MIEP) of human cytomegalovirus (HCMV) controls viral Immediate Early (IE) gene expression, which must be activated to initiate productive infection and repressed to establish latency. Regulation of the MIEP is critical for both viral spread and persistence. In addition to the Daxx-mediated intrinsic cellular defense that regulates the MIEP, the cell-type specific balance between cellular activators and repressors of the promoter may help dictate whether viral IE genes will be expressed or silenced. For example, in undifferentiated myeloid cells, transcriptional repressors of the MIEP may outnumber transcriptional activators, leading to promoter silencing and latency establishment. We created a recombinant viral genome in which a myeloid-active promoter replaced part of the MIEP. The viable virus generated failed to express the viral IE genes in an undifferentiated myeloid cell line. These observations have mechanistic implications regarding how viral IE gene expression is regulated during latency.
Collapse
Affiliation(s)
- Qingsong Qin
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
15
|
Heterologous viral promoters incorporated into the human cytomegalovirus genome are silenced during experimental latency. J Virol 2013; 87:9886-94. [PMID: 23824803 DOI: 10.1128/jvi.01726-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) lytic phase gene expression is repressed upon entry into myeloid lineage cells where the virus establishes latency. Lytic infection is not initiated because the tegument-delivered transactivator protein pp71 fails to enter the nucleus and inactivate the Daxx-mediated cellular intrinsic defense that silences the viral genome. When pp71 is expressed de novo in THP-1 monocytes, it localizes to the nucleus, inactivates the Daxx defense, and initiates lytic infection. We speculated that replacing the native viral promoter that drives pp71 expression with one that is highly and constitutively active in myeloid cells would permit pp71 de novo expression upon infection and that this newly expressed pp71 would accumulate in the nucleus, inactivate the intrinsic defense, and initiate the cascade of lytic gene expression. Surprisingly, we found that this promoter was still subject to normal silencing mechanisms in THP-1 monocytes and primary CD34(+) cells, two independent myeloid lineage cells. A second constitutively active heterologous viral promoter located in a different region of the HCMV genome was also silenced in THP-1 and CD34(+) cells. Furthermore, these two independent heterologous viral promoters inserted into three different regions of the HCMV genome in three different viral strains all required prior expression of the viral immediate early proteins for activation in fibroblasts. From this, we conclude that incorporation within the HCMV genome impacts the proclivity of heterologous viral promoters to initiate transcription. These observations have mechanistic implications for the expression of viral genes and transgenes during both lytic infection and latency.
Collapse
|
16
|
Cao S, Wu C, Yang Y, Sniderhan LF, Maggirwar SB, Dewhurst S, Lu Y. Lentiviral vector-mediated stable expression of sTNFR-Fc in human macrophage and neuronal cells as a potential therapy for neuroAIDS. J Neuroinflammation 2011; 8:48. [PMID: 21569583 PMCID: PMC3118348 DOI: 10.1186/1742-2094-8-48] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/14/2011] [Indexed: 01/23/2023] Open
Abstract
Background Human immunodeficiency virus type 1 (HIV-1) infection frequently causes neurologic disease, which is the result of viral replication and activation of macrophages and microglia in the CNS, and subsequent secretion of high levels of neurotoxic products, including tumor necrosis factor-α (TNF-α). We therefore hypothesized that a soluble TNF-α antagonist might have potential utility as a neuroprotective effecter molecule, and conducted proof-of-concept studies to test this hypothesis. Methods To develop novel therapeutics for the treatment of neuroAIDS, we constructed and characterized a soluble TNF receptor (sTNFR)-Fc fusion protein with the goal of neutralizing TNF-α, and tested the stability of expression of this gene following delivery by a lentiviral vector. Results High-titer lentiviral vectors were prepared, allowing efficient transduction of macrophage/glial and neuronal cell lines, as well as primary rat cerebellar neurons. Efficient, stable secretion of sTNFR-Fc was demonstrated in supernatants from transduced cell lines over 20 passages, using both western blot and ELISA. Biological activity of the secreted sTNFR-Fc was confirmed by TNF-specific in vitro protein binding and functional blocking assays. Finally, the secreted protein was shown to protect neuronal cells from TNF-α, HIV-1 Tat-, and gp120-mediated neurotoxicity. Conclusions These results demonstrate that lentiviral vector mediated expression of sTNFR-Fc may have potential as a novel therapy for neuroAIDS.
Collapse
Affiliation(s)
- Shengbo Cao
- Department of Public Health Sciences, University of Hawai'i, Honolulu, Hawai'i 96822, USA
| | | | | | | | | | | | | |
Collapse
|