235 results on '"Simon Wain-Hobson"'
Search Results
2. Sustained high expression of multiple APOBEC3 cytidine deaminases in systemic lupus erythematosus
- Author
-
Danielle Perez-Bercoff, Hélène Laude, Morgane Lemaire, Oliver Hunewald, Valérie Thiers, Marco Vignuzzi, Hervé Blanc, Aurélie Poli, Zahir Amoura, Vincent Caval, Rodolphe Suspène, François Hafezi, Alexis Mathian, Jean-Pierre Vartanian, and Simon Wain-Hobson
- Subjects
Medicine ,Science - Abstract
Abstract APOBEC3 (A3) enzymes are best known for their role as antiviral restriction factors and as mutagens in cancer. Although four of them, A3A, A3B, A3F and A3G, are induced by type-1-interferon (IFN-I), their role in inflammatory conditions is unknown. We thus investigated the expression of A3, and particularly A3A and A3B because of their ability to edit cellular DNA, in Systemic Lupus Erythematosus (SLE), a chronic inflammatory disease characterized by high IFN-α serum levels. In a cohort of 57 SLE patients, A3A and A3B, but also A3C and A3G, were upregulated ~ 10 to 15-fold (> 1000-fold for A3B) compared to healthy controls, particularly in patients with flares and elevated serum IFN-α levels. Hydroxychloroquine, corticosteroids and immunosuppressive treatment did not reverse A3 levels. The A3AΔ3B polymorphism, which potentiates A3A, was detected in 14.9% of patients and in 10% of controls, and was associated with higher A3A mRNA expression. A3A and A3B mRNA levels, but not A3C or A3G, were correlated positively with dsDNA breaks and negatively with lymphopenia. Exposure of SLE PBMCs to IFN-α in culture induced massive and sustained A3A levels by 4 h and led to massive cell death. Furthermore, the rs2853669 A > G polymorphism in the telomerase reverse transcriptase (TERT) promoter, which disrupts an Ets-TCF-binding site and influences certain cancers, was highly prevalent in SLE patients, possibly contributing to lymphopenia. Taken together, these findings suggest that high baseline A3A and A3B levels may contribute to cell frailty, lymphopenia and to the generation of neoantigens in SLE patients. Targeting A3 expression could be a strategy to reverse cell death and the generation of neoantigens.
- Published
- 2021
- Full Text
- View/download PDF
3. Herpes Simplex Virus Type 1 Infection Disturbs the Mitochondrial Network, Leading to Type I Interferon Production through the RNA Polymerase III/RIG-I Pathway
- Author
-
Noémie Berry, Rodolphe Suspène, Vincent Caval, Pierre Khalfi, Guillaume Beauclair, Stéphane Rigaud, Hervé Blanc, Marco Vignuzzi, Simon Wain-Hobson, and Jean-Pierre Vartanian
- Subjects
HSV-1 ,mitochondria ,innate immunity ,cytidine deaminase ,APOBEC3A ,herpes simplex virus ,Microbiology ,QR1-502 - Abstract
ABSTRACT Viruses have evolved a plethora of mechanisms to impair host innate immune responses. Herpes simplex virus type 1 (HSV-1), a double-stranded linear DNA virus, impairs the mitochondrial network and dynamics predominantly through the UL12.5 gene. We demonstrated that HSV-1 infection induced a remodeling of mitochondrial shape, resulting in a fragmentation of the mitochondria associated with a decrease in their volume and an increase in their sphericity. This damage leads to the release of mitochondrial DNA (mtDNA) to the cytosol. By generating a stable THP-1 cell line expressing the DNase I-mCherry fusion protein and a THP-1 cell line specifically depleted of mtDNA upon ethidium bromide treatment, we showed that cytosolic mtDNA contributes to type I interferon and APOBEC3A upregulation. This was confirmed by using an HSV-1 strain (KOS37 UL98-SPA) with a deletion of the UL12.5 gene that impaired its ability to induce mtDNA stress. Furthermore, by using an inhibitor of RNA polymerase III, we demonstrated that upon HSV-1 infection, cytosolic mtDNA enhanced type I interferon induction through the RNA polymerase III/RIG-I pathway. APOBEC3A was in turn induced by interferon. Deep sequencing analyses of cytosolic mtDNA mutations revealed an APOBEC3A signature predominantly in the 5′TpCpG context. These data demonstrate that upon HSV-1 infection, the mitochondrial network is disrupted, leading to the release of mtDNA and ultimately to its catabolism through APOBEC3-induced mutations. IMPORTANCE Herpes simplex virus 1 (HSV-1) impairs the mitochondrial network through the viral protein UL12.5. This leads to the fusion of mitochondria and simultaneous release of mitochondrial DNA (mtDNA) in a mouse model. We have shown that released mtDNA is recognized as a danger signal, capable of stimulating signaling pathways and inducing the production of proinflammatory cytokines. The expression of the human cytidine deaminase APOBEC3A is highly upregulated by interferon responses. This enzyme catalyzes the deamination of cytidine to uridine in single-stranded DNA substrates, resulting in the catabolism of edited DNA. Using human cell lines deprived of mtDNA and viral strains deficient in UL12, we demonstrated the implication of mtDNA in the production of interferon and APOBEC3A expression during viral infection. We have shown that HSV-1 induces mitochondrial network fragmentation in a human model and confirmed the implication of RNA polymerase III/RIG-I signaling in the capture of cytosolic mtDNA.
- Published
- 2021
- Full Text
- View/download PDF
4. Mouse APOBEC1 cytidine deaminase can induce somatic mutations in chromosomal DNA
- Author
-
Vincent Caval, Wenjuan Jiao, Noémie Berry, Pierre Khalfi, Emmanuelle Pitré, Valérie Thiers, Jean-Pierre Vartanian, Simon Wain-Hobson, and Rodolphe Suspène
- Subjects
APOBEC1 ,Cytidine deaminase ,Somatic mutations ,Nuclear DNA ,Cancer ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background APOBEC1 (A1) enzymes are cytidine deaminases involved in RNA editing. In addition to this activity, a few A1 enzymes have been shown to be active on single stranded DNA. As two human ssDNA cytidine deaminases APOBEC3A (A3A), APOBEC3B (A3B) and related enzymes across the spectrum of placental mammals have been shown to introduce somatic mutations into nuclear DNA of cancer genomes, we explored the mutagenic threat of A1 cytidine deaminases to chromosomal DNA. Results Molecular cloning and expression of various A1 enzymes reveal that the cow, pig, dog, rabbit and mouse A1 have an intracellular ssDNA substrate specificity. However, among all the enzymes studied, mouse A1 appears to be singular, being able to introduce somatic mutations into nuclear DNA with a clear 5’TpC editing context, and to deaminate 5-methylcytidine substituted DNA which are characteristic features of the cancer related mammalian A3A and A3B enzymes. However, mouse A1 activity fails to elicit formation of double stranded DNA breaks, suggesting that mouse A1 possess an attenuated nuclear DNA mutator phenotype reminiscent of human A3B. Conclusions At an experimental level mouse APOBEC1 is remarkable among 12 mammalian A1 enzymes in that it represents a source of somatic mutations in mouse genome, potentially fueling oncogenesis. While the order Rodentia is bereft of A3A and A3B like enzymes it seems that APOBEC1 may well substitute for it, albeit remaining much less active. This modifies the paradigm that APOBEC3 and AID enzymes are the sole endogenous mutator enzymes giving rise to off-target editing of mammalian genomes.
- Published
- 2019
- Full Text
- View/download PDF
5. Pandemic influenza viruses: time to recognize our inability to predict the unpredictable and stop dangerous gain‐of‐function experiments
- Author
-
Simon Wain‐Hobson
- Subjects
gain‐of‐function ,influenza ,pandemic ,vaccines ,virus ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2013
- Full Text
- View/download PDF
6. An Avian H7N1 Gain-of-Function Experiment of Great Concern
- Author
-
Simon Wain-Hobson
- Subjects
Microbiology ,QR1-502 - Abstract
ABSTRACT Inappropriately named gain-of-function influenza research seeks to confer airborne transmission on avian influenza A viruses that otherwise cause only dead-end infections in humans. A recent study has succeeded in doing this with a highly pathogenic ostrich H7N1 virus in a ferret model without loss of virulence. If transposable to humans, this would constitute a novel virus with a case fatality rate ~30 greater than that of Spanish flu. A commentary from three distinguished virologists considered the benefits of this work to outweigh potential risks. I beg to disagree with conclusions in both papers, for the underlying science is not as strong as it appears.
- Published
- 2014
- Full Text
- View/download PDF
7. Human APOBEC3A isoforms translocate to the nucleus and induce DNA double strand breaks leading to cell stress and death.
- Author
-
Bianka Mussil, Rodolphe Suspène, Marie-Ming Aynaud, Anne Gauvrit, Jean-Pierre Vartanian, and Simon Wain-Hobson
- Subjects
Medicine ,Science - Abstract
Human APOBEC3 enzymes deaminate single stranded DNA. At least five can deaminate mitochondrial DNA in the cytoplasm, while three can deaminate viral DNA in the nucleus. However, only one, APOBEC3A, can hypermutate genomic DNA. We analysed the distribution and function of the two APOBEC3A isoforms p1 and p2 in transfected cell lines. Both can translocate to the nucleus and hypermutate CMYC DNA and induce DNA double strand breaks as visualized by the detection of ©H2AX or Chk2. APOBEC3A induced G1 phase cell cycle arrest and triggered several members of the intrinsic apoptosis pathway. Activation of purified human CD4+ T lymphocytes with PHA, IL2 and interferon α resulted in C->T hypermutation of genomic DNA and double stranded breaks suggesting a role for APOBEC3A in pro-inflammatory conditions. As chronic inflammation underlies many diseases including numerous cancers, it is possible that APOBEC3A induction may generate many of the lesions typical of a cancer genome.
- Published
- 2013
- Full Text
- View/download PDF
8. Efficient deamination of 5-methylcytidine and 5-substituted cytidine residues in DNA by human APOBEC3A cytidine deaminase.
- Author
-
Rodolphe Suspène, Marie-Ming Aynaud, Jean-Pierre Vartanian, and Simon Wain-Hobson
- Subjects
Medicine ,Science - Abstract
Deamination of 5-methylcytidine (5MeC) in DNA results in a G:T mismatch unlike cytidine (C) deamination which gives rise to a G:U pair. Deamination of C was generally considered to arise spontaneously. It is now clear that human APOBEC3A (A3A), a polynucleotide cytidine deaminase (PCD) with specificity for single stranded DNA, can extensively deaminate human nuclear DNA. It is shown here that A3A among all human PCDs can deaminate 5-methylcytidine in a variety of single stranded DNA substrates both in vitro and in transfected cells almost as efficiently as cytidine itself. This ability of A3A to accommodate 5-methyl moiety extends to other small and physiologically relevant substituted cytidine bases such as 5-hydroxy and 5-bromocytidine. As 5MeCpG deamination hotspots characterize many genes associated with cancer it is plausible that A3A is a major player in the onset of cancer.
- Published
- 2013
- Full Text
- View/download PDF
9. Evolution of the primate APOBEC3A cytidine deaminase gene and identification of related coding regions.
- Author
-
Michel Henry, Christophe Terzian, Martine Peeters, Simon Wain-Hobson, and Jean-Pierre Vartanian
- Subjects
Medicine ,Science - Abstract
The APOBEC3 gene cluster encodes six cytidine deaminases (A3A-C, A3DE, A3F-H) with single stranded DNA (ssDNA) substrate specificity. For the moment A3A is the only enzyme that can initiate catabolism of both mitochondrial and nuclear DNA. Human A3A expression is initiated from two different methionine codons M1 or M13, both of which are in adequate but sub-optimal Kozak environments. In the present study, we have analyzed the genetic diversity among A3A genes across a wide range of 12 primates including New World monkeys, Old World monkeys and Hominids. Sequence variation was observed in exons 1-4 in all primates with up to 31% overall amino acid variation. Importantly for 3 hominids codon M1 was mutated to a threonine codon or valine codon, while for 5/12 primates strong Kozak M1 or M13 codons were found. Positive selection was apparent along a few branches which differed compared to positive selection in the carboxy-terminal of A3G that clusters with A3A among human cytidine deaminases. In the course of analyses, two novel non-functional A3A-related fragments were identified on chromosome 4 and 8 kb upstream of the A3 locus. This qualitative and quantitative variation among primate A3A genes suggest that subtle differences in function might ensue as more light is shed on this increasingly important enzyme.
- Published
- 2012
- Full Text
- View/download PDF
10. Fitness ranking of individual mutants drives patterns of epistatic interactions in HIV-1.
- Author
-
Javier P Martínez, Gennady Bocharov, Anna Ignatovich, Jochen Reiter, Matthias T Dittmar, Simon Wain-Hobson, and Andreas Meyerhans
- Subjects
Medicine ,Science - Abstract
Fitness interactions between mutations, referred to as epistasis, can strongly impact evolution. For RNA viruses and retroviruses with their high mutation rates, epistasis may be particularly important to overcome fitness losses due to the accumulation of deleterious mutations and thus could influence the frequency of mutants in a viral population. As human immunodeficiency virus type 1 (HIV-1) resistance to azidothymidine (AZT) requires selection of sequential mutations, it is a good system to study the impact of epistasis. Here we present a thorough analysis of a classical AZT-resistance pathway (the 41-215 cluster) of HIV-1 variants by fitness measurements in single round infection assays covering physiological drug concentrations ex vivo. The sign and value of epistasis varied and did not predict the epistatic effect on the mutant frequency. This complex behavior is explained by the fitness ranking of the variants that strongly depends on environmental factors, i.e., the presence and absence of drugs and the host cells used. Although some interactions compensate fitness losses, the observed small effect on the relative mutant frequencies suggests that epistasis might be inefficient as a buffering mechanism for fitness losses in vivo. While the use of epistasis-based hypotheses to make general assumptions on the evolutionary dynamics of viral populations is appealing, our data caution their interpretation without further knowledge on the characteristics of the viral mutant spectrum under different environmental conditions.
- Published
- 2011
- Full Text
- View/download PDF
11. Massive APOBEC3 editing of hepatitis B viral DNA in cirrhosis.
- Author
-
Jean-Pierre Vartanian, Michel Henry, Agnès Marchio, Rodolphe Suspène, Marie-Ming Aynaud, Denise Guétard, Minerva Cervantes-Gonzalez, Carlo Battiston, Vincenzo Mazzaferro, Pascal Pineau, Anne Dejean, and Simon Wain-Hobson
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
DNA viruses, retroviruses and hepadnaviruses, such as hepatitis B virus (HBV), are vulnerable to genetic editing of single stranded DNA by host cell APOBEC3 (A3) cytidine deaminases. At least three A3 genes are up regulated by interferon-alpha in human hepatocytes while ectopic expression of activation induced deaminase (AICDA), an A3 paralog, has been noted in a variety of chronic inflammatory syndromes including hepatitis C virus infection. Yet virtually all studies of HBV editing have confined themselves to analyses of virions from culture supernatants or serum where the frequency of edited genomes is generally low (< or = 10(-2)). We decided to look at the nature and frequency of HBV editing in cirrhotic samples taken during removal of a primary hepatocellular carcinoma. Forty-one cirrhotic tissue samples (10 alcoholic, 10 HBV(+), 11 HBV(+)HCV(+) and 10 HCV(+)) as well as 4 normal livers were studied. Compared to normal liver, 5/7 APOBEC3 genes were significantly up regulated in the order: HCV+/-HBV>HBV>alcoholic cirrhosis. A3C and A3D were up regulated for all groups while the interferon inducible A3G was over expressed in virus associated cirrhosis, as was AICDA in approximately 50% of these HBV/HCV samples. While AICDA can indeed edit HBV DNA ex vivo, A3G is the dominant deaminase in vivo with up to 35% of HBV genomes being edited. Despite these highly deleterious mutant spectra, a small fraction of genomes survive and contribute to loss of HBeAg antigenemia and possibly HBsAg immune escape. In conclusion, the cytokine storm associated with chronic inflammatory responses to HBV and HCV clearly up regulates a number of A3 genes with A3G clearly being a major restriction factor for HBV. Although the mutant spectrum resulting from A3 editing is highly deleterious, a very small part, notably the lightly edited genomes, might help the virus evolve and even escape immune responses.
- Published
- 2010
- Full Text
- View/download PDF
12. Genetic editing of HBV DNA by monodomain human APOBEC3 cytidine deaminases and the recombinant nature of APOBEC3G.
- Author
-
Michel Henry, Denise Guétard, Rodolphe Suspène, Christophe Rusniok, Simon Wain-Hobson, and Jean-Pierre Vartanian
- Subjects
Medicine ,Science - Abstract
Hepatitis B virus (HBV) DNA is vulnerable to editing by human cytidine deaminases of the APOBEC3 (A3A-H) family albeit to much lower levels than HIV cDNA. We have analyzed and compared HBV editing by all seven enzymes in a quail cell line that does not produce any endogenous DNA cytidine deaminase activity. Using 3DPCR it was possible to show that all but A3DE were able to deaminate HBV DNA at levels from 10(-2) to 10(-5)in vitro, with A3A proving to be the most efficient editor. The amino terminal domain of A3G alone was completely devoid of deaminase activity to within the sensitivity of 3DPCR ( approximately 10(-4) to 10(-5)). Detailed analysis of the dinucleotide editing context showed that only A3G and A3H have strong preferences, notably CpC and TpC. A phylogenic analysis of A3 exons revealed that A3G is in fact a chimera with the first two exons being derived from the A3F gene. This might allow co-expression of the two genes that are able to restrict HIV-1Deltavif efficiently.
- Published
- 2009
- Full Text
- View/download PDF
13. Data from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
Purpose: To evaluate CD4+ helper functions and antitumor effect of promiscuous universal cancer peptides (UCP) derived from telomerase reverse transcriptase (TERT).Experimental Design: To evaluate the widespread immunogenicity of UCPs in humans, spontaneous T-cell responses against UCPs were measured in various types of cancers using T-cell proliferation and ELISPOT assays. The humanized HLA-DRB1*0101/HLA-A*0201 transgenic mice were used to study the CD4+ helper effects of UCPs on antitumor CTL responses. UCP-based antitumor therapeutic vaccine was evaluated using HLA-A*0201–positive B16 melanoma that express TERT.Results: The presence of a high number of UCP-specific CD4+ T cells was found in the blood of patients with various types of cancer. These UCP-specific T cells mainly produce IFN-γ and TNF-α. In HLA transgenic mice, UCP vaccinations induced high avidity CD4+ TH1 cells and activated dendritic cells that produced interleukin-12. UCP-based vaccination breaks self-tolerance against TERT and enhances primary and memory CTL responses. Furthermore, the use of UCP strongly improves the efficacy of therapeutic vaccination against established B16-HLA-A*0201 melanoma and promotes tumor infiltration by TERT-specific CD8+ T cells.Conclusions: Our results showed that UCP-based vaccinations strongly stimulate antitumor immune responses and could be used to design efficient immunotherapies in multiple types of cancers. Clin Cancer Res; 18(22); 6284–95. ©2012 AACR.
- Published
- 2023
14. Data from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Purpose:Human telomerase reverse transcriptase (hTERT) is highly expressed in >85% of human tumors and is thus considered as a good tumor-associated antigen candidate for vaccine development. We conducted a phase I study to investigate the safety, tolerability, clinical response, and immunogenicity of INVAC-1, a DNA plasmid encoding a modified hTERT protein in patients with relapsed or refractory solid tumors.Patients and Methods:INVAC-1 was either administered by intradermal route followed by electroporation or by Tropis, a needle-free injection system. Safety and tolerability were monitored by clinical and laboratory assessments. Progression-free survival and overall survival were reported using Kaplan–Meier survival analysis. Immunogenicity was studied by ELISpot, Luminex, and Flow Cytometry.Results:Twenty-six patients were treated with INVAC-1 administered at three dose levels (100, 400, and 800 μg). Vaccination was well tolerated and no dose-limiting toxicity was reported. One treatment-related grade 3 SAE was reported. Fifty-eight percent of patients experienced disease stabilization. PFS was 2.7 months, median OS was 15 months, and 1-year survival was reached for 65% of patients. INVAC-1 vaccination stimulated specific anti-hTERT CD4 T-cell response as well as cytotoxic CD8 T-cell response. No evidence of peripheral vaccine-induced immunosuppression was observed.Conclusions:INVAC-1 vaccination was safe, well tolerated, and immunogenic when administered intradermally at the three tested doses in patients with relapsed or refractory cancers. Disease stabilization was observed for the majority of patients (58%) during the treatment period and beyond.See related commentary by Slingluff Jr, p. 529
- Published
- 2023
15. Supplementary Table 2 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Histological diagnosis, age, number of cycles received and Overall Survival
- Published
- 2023
16. Supplementary Figure 3 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Immunophenotyping of circulating CD4 and CD8 T cells
- Published
- 2023
17. Supplementary Table 3 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Number and percentage of patients reporting studytreatment-related AEs per cycle
- Published
- 2023
18. Supplementary Figure 4 from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
PDF file - 57K, UCPs mediated helper effect on CTLs in other models of vaccinations
- Published
- 2023
19. Supplementary Figure 2 from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
PDF file - 65K, UCP2 and UCP4 peptides increase self/TERT pY572 specific CTL responses
- Published
- 2023
20. Supplementary Figure 5 from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
PDF file - 26K, UCP2 specific CD4 T cells recognize the mTERT-derived counterpart peptide p568
- Published
- 2023
21. Supplementary Table 5 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Clinical features of patients with OS 1 year
- Published
- 2023
22. Supplementary Figure 1 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Flow Chart
- Published
- 2023
23. Supplementary Figure 1 from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
PDF file - 51K, Isolation of UCP4-specific CD4 Th1 cell clone from a cancer patient
- Published
- 2023
24. Supplementary Figure 2 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Kaplan-Meier representation of Overall Survival and Progression Free Survival
- Published
- 2023
25. Data not shown from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
This file compiles all the results identified as "data not shown" in the manuscript
- Published
- 2023
26. Supplementary Figure 3 from Universal Cancer Peptide-Based Therapeutic Vaccine Breaks Tolerance against Telomerase and Eradicates Established Tumor
- Author
-
Olivier Adotévi, Christophe Borg, Pierre Langlade-Demoyen, Eric Tartour, Simon Wain-Hobson, Etienne Daguindau, Federico Sandoval, Bertrand Clerc, Emeline Levionnois, Christelle Liard, Christine Sedlik, Yu Chun Lone, Laurent Beziaud, Charline Vauchy, Yann Godet, and Magalie Dosset
- Abstract
PDF file - 38K, Comparison of T-cell help mediated by UCP2 and a HTLV-1 Tax-derived peptide on self/TERT pY988 specific CTL response
- Published
- 2023
27. Supplementary Table 1 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Flow Cytometry antibody references
- Published
- 2023
28. Supplementary Table 4 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Best Overall Response
- Published
- 2023
29. A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Zineb Ghrieb, Ludovic Doucet, Stéphane Culine, Mara Brizard, Marie-Agnès Dragon Durey, Jacques Medioni, Thierry Huet, Rémy DeFrance, Maria Wehbe, Luis Augusto Teixeira, Julie Garibal, Pierre Langlade-Demoyen, Olivier Adotevi, Jean-Jacques Kiladjian, Elodie Pliquet, Marie Escande, Valérie Doppler, Caroline Laheurte, Stéphane Oudard, and Simon Wain-Hobson
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,ELISPOT ,medicine.medical_treatment ,Immunogenicity ,Immunosuppression ,DNA vaccination ,Vaccination ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Telomerase reverse transcriptase ,business ,Survival analysis - Abstract
Purpose: Human telomerase reverse transcriptase (hTERT) is highly expressed in >85% of human tumors and is thus considered as a good tumor-associated antigen candidate for vaccine development. We conducted a phase I study to investigate the safety, tolerability, clinical response, and immunogenicity of INVAC-1, a DNA plasmid encoding a modified hTERT protein in patients with relapsed or refractory solid tumors. Patients and Methods: INVAC-1 was either administered by intradermal route followed by electroporation or by Tropis, a needle-free injection system. Safety and tolerability were monitored by clinical and laboratory assessments. Progression-free survival and overall survival were reported using Kaplan–Meier survival analysis. Immunogenicity was studied by ELISpot, Luminex, and Flow Cytometry. Results: Twenty-six patients were treated with INVAC-1 administered at three dose levels (100, 400, and 800 μg). Vaccination was well tolerated and no dose-limiting toxicity was reported. One treatment-related grade 3 SAE was reported. Fifty-eight percent of patients experienced disease stabilization. PFS was 2.7 months, median OS was 15 months, and 1-year survival was reached for 65% of patients. INVAC-1 vaccination stimulated specific anti-hTERT CD4 T-cell response as well as cytotoxic CD8 T-cell response. No evidence of peripheral vaccine-induced immunosuppression was observed. Conclusions: INVAC-1 vaccination was safe, well tolerated, and immunogenic when administered intradermally at the three tested doses in patients with relapsed or refractory cancers. Disease stabilization was observed for the majority of patients (58%) during the treatment period and beyond. See related commentary by Slingluff Jr, p. 529
- Published
- 2020
30. A DNA telomerase vaccine for canine cancer immunotherapy
- Author
-
Pascal Clayette, Anne-Sophie Pailhes-Jimenez, Anna Kostrzak, Thomas Bestetti, Marion Julithe, Christine Kreuz, Simon Wain-Hobson, Pierre Langlade-Demoyen, Gabriel Chamel, Jessie Thalmensi, Christelle Liard, Emmanuèle Bourges, Ludovic Bourré, Elodie Pliquet, Marie Escande, Olivier Keravel, and Thierry Huet
- Subjects
DNA vaccine ,0301 basic medicine ,Telomerase ,medicine.medical_treatment ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,cancer ,Telomerase reverse transcriptase ,business.industry ,Electro-Gene-Transfer ,ELISPOT ,Cancer ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Oncology ,canine TERT ,030220 oncology & carcinogenesis ,Cancer research ,immunotherapy ,Cancer vaccine ,business ,Research Paper - Abstract
Telomerase reverse transcriptase (TERT) is highly expressed in more than 90% of canine cancer cells and low to absent in normal cells. Given that immune tolerance to telomerase is easily broken both naturally and experimentally, telomerase is an attractive tumor associated antigen for cancer immunotherapy. Indeed, therapeutic trials using human telomerase peptides have been performed. We have developed an immunogenic yet catalytically inactive human telomerase DNA construct that is in clinical trials with patients presenting solid tumors. Paralleling this human construct, we have developed a canine telomerase DNA vaccine, called pDUV5. When administered intradermally to mice combined with electrogene transfer, pDUV5 induced canine TERT specific cytotoxic T-cells as measured by IFN-γ ELISpot assay. Intradermal vaccination of healthy dogs with 400 μg of pDUV5 generated strong, broad and long lasting TERT specific cellular immune responses. In vitro immunization with cTERT peptides revealed the maintenance of cTERT specific T-cells in PBMCs from tumor bearing dogs showing that this repertoire was not depleted. This study highlights the potential of pDUV5 as a cancer vaccine and supports its evaluation for the treatment of spontaneous canine tumors.
- Published
- 2019
31. Genotoxic stress increases cytoplasmic mitochondrial DNA editing by human APOBEC3 mutator enzymes at a single cell level
- Author
-
Vincent Caval, Bianka Mussil, Jean-Pierre Vartanian, Anne Durandy, Simon Wain-Hobson, Rodolphe Suspène, Rétrovirologie moléculaire - Molecular Retrovirology, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), This work was supported by funds from the Institut Pasteur, Centre National de Recherche Scientifique (CNRS), Institut National du Cancer (S-CR14106 to BM) and OSEO (FUI AAP12 to VC)., We thank Jean-Marc Cavaillon for sepsis samples and dosing IL6 and ICAReB platform for serum samples., Rétrovirologie moléculaire, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris]
- Subjects
CD4-Positive T-Lymphocytes ,Mitochondrial DNA ,viruses ,Cell ,lcsh:Medicine ,Genotoxic Stress ,DNA, Mitochondrial ,Quail ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,0302 clinical medicine ,Cytidine deamination ,Cytidine Deaminase ,medicine ,Animals ,Humans ,APOBEC Deaminases ,lcsh:Science ,Cells, Cultured ,Etoposide ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,lcsh:R ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Fibroblasts ,biochemical phenomena, metabolism, and nutrition ,Mitochondria ,3. Good health ,Cell biology ,medicine.anatomical_structure ,chemistry ,Apoptosis ,Cell culture ,Cytoplasm ,030220 oncology & carcinogenesis ,Dactinomycin ,lcsh:Q ,Single-Cell Analysis ,DNA ,DNA Damage - Abstract
International audience; Human cells are stressed by numerous mechanisms that can lead to leakage of mitochondrial DNA (mtDNA) to the cytoplasm and ultimately apoptosis. this agonist DNA constitutes a danger to the cell and is counteracted by cytoplasmic DNases and APOBEC3 cytidine deamination of DNA. To investigate APOBEC3 editing of leaked mtDNA to the cytoplasm, we performed a PCR analysis of APOBEC3 edited cytoplasmic mtDNA (cymtDNA) at the single cell level for primary CD4 + T cells and the established P2 EBV blast cell line. Up to 17% of primary CD4 + T cells showed signs of APOBEC3 edited cymtDNA with ~50% of all mtDNA sequences showing signs of APOBEC3 editing-between 1500-5000 molecules. Although the P2 cell line showed a much lower frequency of stressed cells, the number of edited mtDNA molecules in such cells was of the same order. Addition of the genotoxic molecules, etoposide or actinomycin D increased the number of cells showing APOBEC3 edited cymtDNA to around 40%. These findings reveal a very dynamic image of the mitochondrial network, which changes considerably under stress. APOBEC3 deaminases are involved in the catabolism of mitochondrial DNA to circumvent chronic immune stimulation triggered by released mitochondrial DNA from damaged cells. The APOBEC3 (A3) locus encodes a series of seven genes encoding six functional endogenous cytidine deam-inases with substrate specificity for single stranded DNA (ssDNA) 1. They leave DNA peppered with uracil residues. This process is referred to as genetic editing as it occurs post replication. A3 enzymes leave a telltale editing signature in DNA: most A3 enzymes preferentially edit a cytidine residue in the context of 5′TpC with the exception of A3G, which prefers 5′CpC dinucleotides 2-6. The antiviral role of these A3 enzymes was initially highlighted by their impact on HIV and HBV replication 6-9 and the fact that several A3 genes can be up-regulated by interferon α 10-12. A3 can restrict the transposition of SINE and LINE retroelements 13,14 and mitochondrial DNA (mtDNA) in cell lines and tissues, all of which show the A3 editing signature-5′TpC and 5′CpC are preferentially deam-inated 15. For the latter, the target was cytoplasmic mtDNA (cymtDNA) in keeping with the observation that all A3 enzymes are unable to access the mitochondrial network 15. In addition, A3A and to a lesser extent A3B, were shown to target chromosomal DNA non-specifically, leading to hypermutated DNA with up to 70% of cyti-dine residues deaminated 15,16. A3A and A3B-induced editing occur predominantly on the lagging strand during DNA replication 17,18. Interestingly, A3A and A3B can edit 5-methylcytidine (5MeC) residues in ssDNA which makes sense given that 5MeC is found exclusively in the nucleus 16,19-21. Uracil bases in DNA are excised by uracil N-glycosidase (UNG) that initiates DNA damage responses, which can result in either DNA repair or catabolism.
- Published
- 2019
32. Sustained high expression of multiple APOBEC3 cytidine deaminases in systemic lupus erythematosus
- Author
-
Valérie Thiers, Aurélie Poli, Vincent Caval, François Hafezi, Morgane Lemaire, Hélène Laude, Hervé Blanc, Jean-Pierre Vartanian, Marco Vignuzzi, Danielle Perez-Bercoff, Simon Wain-Hobson, Alexis Mathian, Rodolphe Suspène, Zahir Amoura, Oliver Hunewald, Gestionnaire, Hal Sorbonne Université, Luxembourg Institute of Health (LIH), Investigation Clinique et d’Accès aux Ressources Biologiques (Plate-forme) - Clinical Investigation and Access to BioResources (ICAReB), Institut Pasteur [Paris] (IP), Populations virales et Pathogenèse - Viral Populations and Pathogenesis, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Génétique Moléculaire des Virus à ARN - Molecular Genetics of RNA Viruses (GMV-ARN (UMR_3569 / U-Pasteur_2)), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Institut E3M [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Centre d'Immunologie et des Maladies Infectieuses (CIMI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Service de Département de médecine interne et immunologie clinique [CHU Pitié-Salpêtrière] (DMIIC), Centre National de Référence du Lupus Systémique, Syndrome des Anticorps Anti-phospholipides et Maladies Auto-immunes Systémiques Rares [CHU Pitié Salpêtrière], Service de Médecine Interne 2, maladies auto-immunes et systémiques [CHU Pitié-Salpêtrière], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Institut E3M [CHU Pitié-Salpêtrière], This work was supported by Fonds National de la Recherche du Luxembourg (INTER/Mobility/16/1154722/APO-SLE), by Ministère de l’Education et de la Recherche du Luxembourg and by Institut Pasteur Paris. FH was supported by FNR-PRIDE scheme (PRIDE/11012546/NEXTIMMUNE)., Institut Pasteur [Paris], Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris]-Université de Paris (UP), Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Centre d'Immunologie et de Maladies Infectieuses (CIMI), Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut E3M [CHU Pitié-Salpêtrière], Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Service de médecine interne et d'immunologie clinique [CHU Pitié-Salpêtrière], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)
- Subjects
Adult ,Male ,Programmed cell death ,Science ,[SDV]Life Sciences [q-bio] ,Cell ,Immunology ,Peripheral blood mononuclear cell ,Polymorphism, Single Nucleotide ,Gene Expression Regulation, Enzymologic ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Polymorphism (computer science) ,Genetics ,Medicine ,Humans ,Lupus Erythematosus, Systemic ,Telomerase reverse transcriptase ,APOBEC Deaminases ,Telomerase ,Germ-Line Mutation ,030304 developmental biology ,Cancer ,0303 health sciences ,Multidisciplinary ,Cell Death ,business.industry ,Interferon-alpha ,Hydroxychloroquine ,medicine.disease ,3. Good health ,Up-Regulation ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
APOBEC3 (A3) enzymes are best known for their role as antiviral restriction factors and as mutagens in cancer. Although four of them, A3A, A3B, A3F and A3G, are induced by type-1-interferon (IFN-I), their role in inflammatory conditions is unknown. We thus investigated the expression of A3, and particularly A3A and A3B because of their ability to edit cellular DNA, in Systemic Lupus Erythematosus (SLE), a chronic inflammatory disease characterized by high IFN-α serum levels. In a cohort of 57 SLE patients, A3A and A3B, but also A3C and A3G, were upregulated ~ 10 to 15-fold (> 1000-fold for A3B) compared to healthy controls, particularly in patients with flares and elevated serum IFN-α levels. Hydroxychloroquine, corticosteroids and immunosuppressive treatment did not reverse A3 levels. The A3AΔ3B polymorphism, which potentiates A3A, was detected in 14.9% of patients and in 10% of controls, and was associated with higher A3A mRNA expression. A3A and A3B mRNA levels, but not A3C or A3G, were correlated positively with dsDNA breaks and negatively with lymphopenia. Exposure of SLE PBMCs to IFN-α in culture induced massive and sustained A3A levels by 4 h and led to massive cell death. Furthermore, the rs2853669 A > G polymorphism in the telomerase reverse transcriptase (TERT) promoter, which disrupts an Ets-TCF-binding site and influences certain cancers, was highly prevalent in SLE patients, possibly contributing to lymphopenia. Taken together, these findings suggest that high baseline A3A and A3B levels may contribute to cell frailty, lymphopenia and to the generation of neoantigens in SLE patients. Targeting A3 expression could be a strategy to reverse cell death and the generation of neoantigens.
- Published
- 2021
33. Luc Montagnier (1932–2022)
- Author
-
Robin A, Weiss and Simon, Wain-Hobson
- Subjects
Multidisciplinary ,viruses - Abstract
Discoverer of the human immunodeficiency virus
- Published
- 2022
34. SOUS-CHAPITRE 9.1 Les défis de la microbiologie fondamentale : connaissance du monde microbien, génomique, physiologie-métabolisme, parois
- Author
-
Simon Wain-Hobson and Bernard Dujon
- Published
- 2020
35. Elephant APOBEC3A cytidine deaminase induces massive double-stranded DNA breaks and apoptosis
- Author
-
Vincent Caval, Xiongxiong Li, Jean-Pierre Vartanian, Simon Wain-Hobson, Rétrovirologie Moléculaire, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Lanzhou Institute of Biological Products Co. Ltd. (LIBP), This work was supported by funds from the Institut Pasteur, the Centre National de la Recherche Scientifique (CNRS) and the Association pour la Recherche sur le Cancer (ARC, S-CR16140). XXL was supported by a stipend from Lanzhou Institute of Biological Products Co., Ltd (LIBP), subsidiary company of China National Biotec Group Company Limited (CNBG) and by a stipend from the Pasteur-Paris University (PPU) International PhD program, We would like to thank Drs Aurore Romey, Sandra Blaise-Boisseau and Labib Bakkali Kassimi for providing elephant tissues and for RNA extraction., and Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Somatic cell ,Elephants ,lcsh:Medicine ,Apoptosis ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Cytidine ,Biology ,Genome ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cytidine Deaminase ,Animals ,Humans ,DNA Breaks, Double-Stranded ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,APOBEC3A ,lcsh:Science ,Gene ,Genetics ,Multidisciplinary ,Body Weight ,lcsh:R ,Cytidine deaminase ,genomic DNA ,030104 developmental biology ,chemistry ,Mutagenesis ,Mutation ,lcsh:Q ,030217 neurology & neurosurgery ,DNA ,HeLa Cells - Abstract
International audience; The incidence of developing cancer should increase with the body mass, yet is not the case, a conundrum referred to as Peto's paradox. Elephants have a lower incidence of cancer suggesting that these animals have probably evolved different ways to protect themselves against the disease. The paradox is worth revisiting with the realization that most mammals encode an endogenous APOBEC3 cytidine deaminase capable of mutating single stranded DNA. Indeed, the mutagenic activity of some APOBEC3 enzymes has been shown to introduce somatic mutations into genomic DNA. These enzymes are now recognized as causal agent responsible for the accumulation of CG-> TA transitions and DNA breaks leading to chromosomal rearrangements in human cancer genomes. Here, we identified an elephant A3Z1 gene, related to human APOBEC3A and showed that it could efficiently deaminate cytidine, 5-methylcytidine and produce DNA breaks leading to massive apoptosis, similar to other mammalian APOBEC3A enzymes where body mass varies by up to four orders of magnitude. Consequently, it could be considered that eAZ1 might contribute to cancer in elephants in a manner similar to their proposed role in humans. If so, eAZ1 might be particularly well regulated to counter Peto's paradox. The APOBEC3 (A3) locus is bounded by two conserved genes, chromobox 6 and 7 (CBX6 and CBX7) in most pla-cental mammals and encodes a family of cytidine deaminases capable of converting cytidine residues to uridine in single strand DNA (ssDNA). The mutagenic activity of these enzymes is involved with the restriction of retro-viruses and DNA viruses, as well as endogenous retroelements and retrotransposons through hypermutation of viral DNA in a process called editing 1. The A3 repertoire is extremely variable among mammals, the locus being shaped through extensive gene duplications and functionalization in the context of a virus-host arms race. A3 enzymes are made up of three related, but distinct zinc-finger domains referred to as Z1, Z2 and Z3 2-4 presumably already present in the genome of the ancestor of placental mammals 5. The last few years has seen the identification of two human endogenous A3 cytidine deaminases, APOBEC3A (A3A) and APOBEC3B (A3B) capable of introducing multiple mutations in chromosomal DNA 6-9. These findings are grounded by the analysis of many cancer genomes, revealing far more mutations and rearrangements than hitherto imagined, where CG-> TA transitions appears to be the dominant mutations 10-13. Human A3A is composed of a single Z1 domain, while A3B is composed of a double Z2Z1 domain, although only the C terminal Z1 domain being catalytically functional 6. A3A and A3B enzymes are both localized in the nucleus and can edit cytidine residues to uridine in ssDNA during transcription and replication, following DNA repair, and leave TpC to TpT signature mutations that show up in cancer genomes 6,8,9. Both enzymes can mutate 5-methylcytidine (5MeC) to thymidine leaving another distinct signature in cancer genomes 6,14-16. Although A3A and A3B are accepted as intrinsic mutators of cellular chromosomal DNA, analyzed in several cancer types 8,11,17 , debate still persists regarding the contribution of each enzyme in the accumulation of mutations paving the way for oncogenesis. While, it has been described that A3A and A3B could be enzymat-ically active in different cancers 18 , A3A is the more active of the two enzymes and as a consequence, only A3A can produce double stranded breaks (DSBs), at least in an experimental setting 6,7,19. Editing frequencies of >0.5 can be found which is why the phenomenon is referred to as hyperediting or hypermutation 9. Accumulation of substitutions localized in the A3B C-terminal domain attenuated the activity of the enzyme compared to A3A 6 .
- Published
- 2019
36. Mouse APOBEC1 cytidine deaminase can induce somatic mutations in chromosomal DNA
- Author
-
Pierre Khalfi, Valérie Thiers, Jean-Pierre Vartanian, Noémie Berry, Wenjuan Jiao, Vincent Caval, Rodolphe Suspène, Emmanuelle Pitré, Simon Wain-Hobson, Rétrovirologie moléculaire, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Université Paris Diderot - Paris 7 (UPD7), Université Sorbonne Paris Cité (USPC), ED 515 - Complexité du vivant, Sorbonne Université (SU), This work was supported by grants from the Institut Pasteur and Centre National de la Recherche Scientifique (CNRS). NB and EP were supported by Allocations de Recherche du Ministère de la Recherche while PK was supported by La Ligue contre le Cancer. WJ was supported by China Scholarship Council., We would like to thank Yu Wei for mouse samples., Rétrovirologie moléculaire - Molecular Retrovirology, and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH: DNA Breaks, Double-Stranded ,Gene Expression ,MESH: Amino Acid Sequence ,MESH: Base Sequence ,medicine.disease_cause ,Substrate Specificity ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,DNA Breaks, Double-Stranded ,MESH: Animals ,APOBEC3A ,MESH: Phylogeny ,Phylogeny ,Cancer ,0303 health sciences ,Nuclear DNA ,Cytidine ,Cytidine deaminase ,APOBEC1 ,Cell biology ,MESH: DNA, Single-Stranded ,RNA editing ,030220 oncology & carcinogenesis ,Biotechnology ,Research Article ,MESH: Enzyme Activation ,MESH: Gene Expression ,lcsh:QH426-470 ,lcsh:Biotechnology ,APOBEC-1 Deaminase ,MESH: APOBEC-1 Deaminase/metabolism ,DNA, Single-Stranded ,Molecular cloning ,Biology ,MESH: APOBEC-1 Deaminase/chemistry ,03 medical and health sciences ,Somatic mutations ,lcsh:TP248.13-248.65 ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,medicine ,Animals ,MESH: RNA Editing ,Amino Acid Sequence ,MESH: Mice ,030304 developmental biology ,Base Sequence ,MESH: Chromosomes, Mammalian/genetics ,MESH: APOBEC-1 Deaminase/genetics ,Chromosomes, Mammalian ,Enzyme Activation ,lcsh:Genetics ,chemistry ,Mutation ,MESH: Mutation ,MESH: Substrate Specificity ,RNA Editing ,Carcinogenesis ,DNA - Abstract
BackgroundAPOBEC1 (A1) enzymes are cytidine deaminases involved in RNA editing. In addition to this activity, a few A1 enzymes have been shown to be active on single stranded DNA. As two human ssDNA cytidine deaminases APOBEC3A (A3A), APOBEC3B (A3B) and related enzymes across the spectrum of placental mammals have been shown to introduce somatic mutations into nuclear DNA of cancer genomes, we explored the mutagenic threat of A1 cytidine deaminases to chromosomal DNA.ResultsMolecular cloning and expression of various A1 enzymes reveal that the cow, pig, dog, rabbit and mouse A1 have an intracellular ssDNA substrate specificity. However, among all the enzymes studied, mouse A1 appears to be singular, being able to introduce somatic mutations into nuclear DNA with a clear 5’TpC editing context, and to deaminate 5-methylcytidine substituted DNA which are characteristic features of the cancer related mammalian A3A and A3B enzymes. However, mouse A1 activity fails to elicit formation of double stranded DNA breaks, suggesting that mouse A1 possess an attenuated nuclear DNA mutator phenotype reminiscent of human A3B.ConclusionsAt an experimental level mouse APOBEC1 is remarkable among 12 mammalian A1 enzymes in that it represents a source of somatic mutations in mouse genome, potentially fueling oncogenesis. While the orderRodentiais bereft of A3A and A3B like enzymes it seems that APOBEC1 may well substitute for it, albeit remaining much less active. This modifies the paradigm that APOBEC3 and AID enzymes are the sole endogenous mutator enzymes giving rise to off-target editing of mammalian genomes.
- Published
- 2019
37. Hypoxia-induced human deoxyribonuclease I is a cellular restriction factor of hepatitis B virus
- Author
-
Camille Hallez, Cristina M. Dorobantu, Mohamed S Bouzidi, Valérie Thiers, Vincent Lucansky, Jean-Pierre Vartanian, Xiongxiong Li, Simon Wain-Hobson, Raphael Gaudin, Rodolphe Suspène, Rétrovirologie moléculaire - Molecular Retrovirology, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Collège Doctoral, Sorbonne Université (SU), China National Biotec Group (CNBG), Institut de Recherche sur les Maladies Virales et Hépatiques (IVH), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM), Viroclinics Biosciences B. V. [Rotterdam, Netherlands], Biomedical Center Martin [Slovaquie], Jessenius Medical Faculty [Commenius University, Slovaquie] (JFMED), Commenius University in Bratislava - Univerzita Komenského-Commenius University in Bratislava - Univerzita Komenského, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), This work was supported by grants from the Institut Pasteur and Centre National de la Recherche Scientifique. C.H. and M.S.B. were supported by the Allocation de Recherche du Ministère de la Recherche and Ligue contre le Cancer, respectively. X.L. was supported by a stipend from the Lanzhou Institute of Biological Products Co., Ltd, subsidiary company of China National Biotec Group Company Limited and by a stipend from the Pasteur-Paris University International PhD program. V.L. was supported by the Fondation pour la Recherche Medicale (project no. ING20160435328). C.D. was supported by an EMBO Long Term Fellowship (EMBO ALTF 1428–2016). The study of patients was sponsored by the French National Agency for research on AIDS and hepatitis (ANRS). This work has been published within the framework of IdEx Université de Strasbourg and has received funding from the French State via the French National Research Agency (ANR) as part of the program ‘Investissements d’avenir’ to R.G. This work was supported by an ATIP-AVENIR starting grant to R.G., We would like to thank S. Urban for providing HepG2-NTCP cells, P. Pineau and A. Marchio for the cirrhotic liver samples, M.-L. Michel and S. Pol for serum samples, C. Neuveut, P. Moreau, P. Maillard and B. Quioc for HBV infection, Y. Jacob for the HIF-1α and HIF-1β plasmids, J. Krijnse-Locker and C. Schmitt for electron microscopy immunolabelling and V. Caval, N. Frampton and J. McKeating for helpful discussions., Rétrovirologie moléculaire, Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], Collège doctoral [Sorbonne universités], Vartanian, Jean-Pierre, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), and Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)
- Subjects
Liver Cirrhosis ,viruses ,[SDV]Life Sciences [q-bio] ,MESH: Virus Replication*/drug effects ,Gene Expression ,medicine.disease_cause ,Virus Replication ,Applied Microbiology and Biotechnology ,chemistry.chemical_compound ,Hypoxia ,ComputingMilieux_MISCELLANEOUS ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,0303 health sciences ,MESH: Liver Cirrhosis/enzymology ,Cytidine ,Transfection ,Cobalt ,Hepatitis B ,MESH: Cobalt/pharmacology ,MESH: Deoxyribonuclease I/metabolism ,Hepatitis B Core Antigens ,3. Good health ,[SDV] Life Sciences [q-bio] ,Capsid ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Hypoxia-Inducible Factor 1 ,MESH: Deoxyribonuclease I/genetics ,Microbiology (medical) ,Hepatitis B virus ,MESH: Mutation ,MESH: Gene Expression ,MESH: Hepatitis B/enzymology ,MESH: Hypoxia-Inducible Factor 1/metabolism ,Immunology ,Biology ,Microbiology ,Gene Expression Regulation, Enzymologic ,Cell Line ,03 medical and health sciences ,Genetics ,medicine ,Deoxyribonuclease I ,Humans ,MESH: Gene Expression Regulation, Enzymologic/drug effects ,030304 developmental biology ,MESH: Hypoxia*/chemically induced ,MESH: Humans ,030306 microbiology ,Virion ,MESH: Virion/metabolism ,MESH: Hepatitis B Core Antigens/metabolism ,Cell Biology ,medicine.disease ,Molecular biology ,In vitro ,MESH: Cell Line ,MESH: DNA, Viral/metabolism ,chemistry ,DNA, Viral ,Mutation ,DNA ,MESH: Hepatitis B virus/physiology - Abstract
International audience; Numerous human APOBEC3 cytidine deaminases have proven to be, inter alia, host cell restriction factors for retroviruses and hepadnaviruses. Although they can bind to genomic RNA and become encapsidated, they are only catalytically active on single-stranded DNA. As there are many cellular deoxyribonucleases (DNases), we hypothesized that a parallel could be struck between APOBEC3 and DNases. For human hepatitis B virus (HBV), we show that DNase I can considerably reduce the virion genome copy number from a variety of transfected or infected cells. DNASE1 is overexpressed and encapsidated in HBV particles in vitro in hypoxic environments and in vivo in cirrhotic patient livers as well as in the serum of infected patients. The use of CoCl2 and dimethyloxalylglycine, mimetic agents used to induce hypoxia by inhibiting prolyl hydroxylase enzymes that stabilize hypoxia-inducible factor (HIF)-1α, showed that the formation of HIF-1α/HIF-1β heterodimers results in the induction of DNASE1. Indeed, transfection with HIF-1α and HIF-1β expression constructs upregulated DNASE1. These findings suggest that human DNase I can impact HBV replication through the catabolism of the DNA genome within the capsid. The activity of DNases in general may explain in part the high frequency of empty or 'light' hepatitis B virions observed in vivo.
- Published
- 2019
38. Strong antigen-specific T-cell immunity induced by a recombinant human TERT measles virus vaccine and amplified by a DNA/viral vector prime boost in IFNAR/CD46 mice
- Author
-
Frédéric Tangy, Thierry Huet, Pierre Langlade-Demoyen, Christelle Liard, Marion Julithe, Marie Escande, Chantal Combredet, Jessie Thalmensi, Simon Wain-Hobson, Elodie Pliquet, Claude Ruffié, Thomas Bestetti, Valérie Najburg, Invectys, BioTop Institut Pasteur, Rétrovirologie Moléculaire, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Génomique virale et vaccination, Funding was provided by Association Nationale de la Recherche et de la Technologie (Grant no. 2012/0152).We would like to thank Shannon A. Fairbanks and Bernardo Fort Brescia for their unswerving support. Elodie Pliquet was supported by an industrial Ph.D. fellowship from the French National Association of Research and Technology (ANRT)., The authors would like to thank Ludovic Bourré, Anne-Sophie Pailhes-Jimenez, Emanuèle Bourges, Rahima Youssi and Pascal Clayette for experimental and editorial help, and the staff of the Institut Pasteur’s animal facilities and imaging platform, and Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Cancer Research ,T-Lymphocytes ,medicine.medical_treatment ,Epitopes, T-Lymphocyte ,Priming (immunology) ,CD8-Positive T-Lymphocytes ,MESH: Cancer Vaccines ,law.invention ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,law ,MESH: Genetic Vectors ,MESH: Chlorocebus aethiops ,Chlorocebus aethiops ,Vaccines, DNA ,Immunology and Allergy ,MESH: Animals ,Telomerase ,Cancer ,Immunity, Cellular ,MESH: Cytokines ,biology ,MESH: CD4-Positive T-Lymphocytes ,MESH: Telomerase ,MESH: CD8-Positive T-Lymphocytes ,3. Good health ,Vaccination ,Oncology ,Recombinant DNA ,Cytokines ,MESH: Immunization ,Immunotherapy ,hTERT ,MESH: Mice, Transgenic ,Genetic Vectors ,Immunology ,Immunization, Secondary ,MESH: Vero Cells ,Mice, Transgenic ,Cancer Vaccines ,Cell Line ,Viral vector ,DNA vaccination ,MESH: Immunity, Cellular ,Measles virus ,MESH: Epitopes, T-Lymphocyte ,03 medical and health sciences ,Immune system ,Measles virus vaccine ,medicine ,Animals ,Humans ,MESH: Immunization, Secondary ,MESH: Cytotoxicity, Immunologic ,Vero Cells ,MESH: Mice ,MESH: Humans ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,biology.organism_classification ,Virology ,MESH: Vaccines, DNA ,MESH: Cell Line ,MESH: T-Lymphocytes ,T-cell responses ,Immunization ,[SDV.IMM.VAC]Life Sciences [q-bio]/Immunology/Vaccinology ,MESH: Measles virus ,030215 immunology - Abstract
International audience; Cancer immunotherapy is seeing an increasing focus on vaccination with tumor-associated antigens (TAAs). Human telomerase (hTERT) is a TAA expressed by most tumors to overcome telomere shortening. Tolerance to hTERT can be easily broken both naturally and experimentally and hTERT DNA vaccine candidates have been introduced in clinical trials. DNA prime/boost strategies have been widely developed to immunize efficiently against infectious diseases. We explored the use of a recombinant measles virus (MV) hTERT vector to boost DNA priming as recombinant live attenuated measles virus has an impressive safety and efficacy record. Here, we show that a MV-TERT vector can rapidly and strongly boost DNA hTERT priming in MV susceptible IFNAR/CD46 mouse models. The cellular immune responses were Th1 polarized. No humoral responses were elicited. The 4 kb hTERT transgene did not impact MV replication or induction of cell-mediated responses. These findings validate the MV-TERT vector to boost cell-mediated responses following DNA priming in humans.
- Published
- 2019
39. The rabbit as an orthologous small animal model for APOBEC3A oncogenesis
- Author
-
Michel Henry, Mohamed S. Bouzidi, Xiongxiong Li, Hélène Laude, Vincent Caval, Florence Jamet, Rodolphe Suspène, Jean-Pierre Vartanian, Simon Wain-Hobson, Rétrovirologie moléculaire, Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], Lanzhou Institute of Biological Products Co. Ltd. (LIBP), This work was supported by funds from the Institut Pasteur and Centre National de la Recherche Scientifique (CNRS). Hélène Laude, Vincent Caval and Mohamed Salah Bouzidi were supported by the 'Assistance Publique des Hôpitaux de Paris', OSEO (FUI AAP12), and the Ligue Nationale contre le Cancer (GB/MA/CD-11283) respectively, Xiong Xiong Li was supported by a stipend from Lanzhou Institute of Biological Products Co., Ltd (LIBP), subsidiary company of China National Biotec Group Company Limited (CNBG) and by a stipend from the Pasteur - Paris University (PPU) International PhD program., We thank Dr. Robin Lombard from Miltenyi Biotec for his help in setting up the FRET-FACS assay, Rétrovirologie moléculaire - Molecular Retrovirology, and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,APOBEC3A Gene ,rabbit ,Locus (genetics) ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,cancer ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,APOBEC3A ,Gene ,cytidine deaminase ,Genetics ,030102 biochemistry & molecular biology ,animal model ,Cytidine ,Cytidine deaminase ,3. Good health ,Nuclear DNA ,genomic DNA ,030104 developmental biology ,Oncology ,chemistry ,Research Paper - Abstract
// Helene C. Laude 1 , Vincent Caval 1 , Mohamed S. Bouzidi 1 , Xiongxiong Li 1, 2 , Florence Jamet 1 , Michel Henry 1 , Rodolphe Suspene 1 , Simon Wain-Hobson 1 and Jean-Pierre Vartanian 1 1 Molecular Retrovirology Unit, Institut Pasteur, CNRS UMR 3569, France 2 Lanzhou Institute of Biological Products Co., Ltd (LIBP), subsidiary company of China National Biotec Group Company Limited (CNBG), Lanzhou 730046, China Correspondence to: Jean-Pierre Vartanian, email: jean-pierre.vartanian@pasteur.fr Keywords: rabbit; animal model; APOBEC3A; cytidine deaminase; cancer Received: March 15, 2018 Accepted: May 24, 2018 Published: June 12, 2018 ABSTRACT APOBEC3 are cytidine deaminases that convert cytidine to uridine residues. APOBEC3A and APOBEC3B enzymes able to target genomic DNA are involved in oncogenesis of a sizeable proportion of human cancers. While the APOBEC3 locus is conserved in mammals, it encodes from 1–7 genes. APOBEC3A is conserved in most mammals, although absent in pigs, cats and throughout Rodentia whereas APOBEC3B is restricted to the Primate order. Here we show that the rabbit APOBEC3 locus encodes two genes of which APOBEC3A enzyme is strictly orthologous to human APOBEC3A. The rabbit enzyme is expressed in the nucleus and the cytoplasm, it can deaminate cytidine, 5-methcytidine residues, nuclear DNA and induce double-strand DNA breaks. The rabbit APOBEC3A enzyme is negatively regulated by the rabbit TRIB3 pseudokinase protein which is guardian of genome integrity, just like its human counterpart. This indicates that the APOBEC3A/TRIB3 pair is conserved over approximately 100 million years. The rabbit APOBEC3A gene is widely expressed in rabbit tissues, unlike human APOBEC3A . These data demonstrate that rabbit could be used as a small animal model for studying APOBEC3 driven oncogenesis.
- Published
- 2018
40. Plurality in HIV Genetics
- Author
-
Simon Wain-Hobson
- Subjects
Genetics ,business.industry ,Human immunodeficiency virus (HIV) ,medicine ,medicine.disease_cause ,business - Published
- 2018
41. APOBEC3A intratumoral DNA electroporation in mice
- Author
-
Vincent Caval, J Thalmensi, T Bestetti, E Pliquet, L Fiette, M Julithe, T. Huet, Anna Kostrzak, M Escande, Simon Wain-Hobson, Pierre Langlade-Demoyen, Invectys [Paris], Rétrovirologie moléculaire, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Histopathologie humaine et Modèles animaux, Institut Pasteur [Paris], Université Sorbonne Paris Cité (USPC), Département Infection et Epidémiologie - Department of Infection and Epidemiology, This work was supported by Invectys., Rétrovirologie moléculaire - Molecular Retrovirology, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), and Institut Pasteur [Paris] (IP)
- Subjects
0301 basic medicine ,Male ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Cancer therapy ,Hydrolases ,Genetic enhancement ,[SDV]Life Sciences [q-bio] ,MESH: Electroporation/methods ,Melanoma, Experimental ,Mice ,0302 clinical medicine ,Tumor Cells, Cultured ,MESH: Animals ,Fibrosarcoma ,Melanoma ,MESH: Lymphocytes, Tumor-Infiltrating/immunology ,MESH: Cytidine Deaminase/genetics ,Electroporation ,MESH: Melanoma, Experimental/therapy ,Cytidine deaminase ,Cytological techniques ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Plasmids ,Somatic hypermutation ,Vectors in gene therapy ,Gene delivery ,Biology ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,MESH: Melanoma, Experimental/genetics ,MESH: Mice, Inbred C57BL ,Cytidine Deaminase ,Genetics ,medicine ,MESH: Genetic Therapy ,Animals ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,MESH: Tumor Cells, Cultured ,Molecular Biology ,MESH: Mice ,Proteins ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Genetic Therapy ,medicine.disease ,Molecular biology ,MESH: Male ,Mice, Inbred C57BL ,030104 developmental biology ,MESH: Proteins/genetics ,Genetic engineering ,MESH: Melanoma, Experimental/immunology ,MESH: Plasmids/genetics ,MESH: Female - Abstract
International audience; Human APOBEC3A (A3A) cytidine deaminase shows pro-apoptotic properties resulting from hypermutation of genomic DNA, induction of double-stranded DNA breaks (DSBs) and G1 cell cycle arrest. Given this, we evaluated the antitumor efficacy of A3A by intratumoral electroporation of an A3A expression plasmid. DNA was repeatedly electroporated into B16OVA, B16Luc tumors of C57BL/6J mice as well as the aggressive fibrosarcoma Sarc2 tumor of HLA-A*0201/DRB1*0101 transgenic mice using noninvasive plate electrodes. Intratumoral electroporation of A3A plasmid DNA resulted in regression of ~50% of small B16OVA melanoma tumors that did not rebound in the following 2 months without treatment. Larger or more aggressive tumors escaped regression when so treated. As APOBEC3A was much less efficient in provoking hypermutation and DSBs in B16OVA cells compared with human or quail cells, it is likely that APOBEC3A would be more efficient in a human setting than in a mouse model.
- Published
- 2017
42. APOBEC3A catabolism of electroporated plasmid DNA in mouse muscle
- Author
-
Anna Kostrzak, Jean-Pierre Vartanian, Demoyen Pl, Michel Henry, and Simon Wain-Hobson
- Subjects
DNA repair ,DNA polymerase II ,Molecular Sequence Data ,Biology ,Molecular cloning ,Transfection ,DNA, Mitochondrial ,DNA polymerase delta ,Cell Line ,Electron Transport Complex IV ,Mutation Rate ,Genes, Reporter ,Cytidine Deaminase ,Genetics ,Animals ,DNA Cleavage ,Luciferases ,Muscle, Skeletal ,Molecular Biology ,Cell Nucleus ,Mice, Inbred BALB C ,DNA clamp ,Base Sequence ,Circular bacterial chromosome ,Proteins ,Electroporation ,Biochemistry ,Uracil-DNA glycosylase ,Mutation ,biology.protein ,Molecular Medicine ,Female ,In vitro recombination ,Plasmids - Abstract
The mouse is widely used as a model for DNA therapy and vaccination even though the efficiency of DNA delivery in higher mammals and humans is much less. The human APOBEC3 (A3) enzymes impact viral genomes by cytidine deamination, which introduces multiple uridine residues into single-stranded DNA, a process known as genetic editing. This initiates rapid DNA catabolism via a uracil DNA glycosylase dependent pathway. In tissue culture, A3A, A3C and A3B can hyperedit transfected plasmid DNA. We explored plasmid catabolism in vivo initiated by A3A, the most efficient of the human enzymes and one that is functionally conserved across most mammals. As rodents do not encode an A3A enzyme, it was possible to explore DNA degradation in the mouse model. Human A3A genetically edits co-electroporated luciferase plasmid DNA in mouse skeletal muscle that initiates DNA degradation resulting in approximately fourfold decrease in bioluminescence. Part of the degradation occurs in the nucleus as indicated by complex hyperedited DNA molecules. As human A3A is strongly upregulated by interferon α and DNA sensing pathways, it is a strong candidate enzyme for restricting plasmid DNA in higher mammals.
- Published
- 2014
43. Erroneous identification of APOBEC3-edited chromosomal DNA in cancer genomics
- Author
-
Caval, Mohamed S. Bouzidi, Jean-Pierre Vartanian, Michel Henry, Rodolphe Suspène, Simon Wain-Hobson, Rétrovirologie Moléculaire, Institut Pasteur [Paris] (IP), and This work was supported by grants from the Institut Pasteur, INCa and the CNRS. VC and MSB were supported by OSEO and the Ligue Nationale Contre le Cancer.
- Subjects
MESH: Neoplasm Proteins ,CD4-Positive T-Lymphocytes ,Cancer Research ,[SDV]Life Sciences [q-bio] ,MESH: beta Catenin ,Cytidine ,medicine.disease_cause ,Nucleic Acid Denaturation ,Genome ,Polymerase Chain Reaction ,law.invention ,Cytosine Deaminase ,chemistry.chemical_compound ,MESH: Liver Neoplasms ,law ,APOBEC Deaminases ,APOBEC3A ,MESH: Carcinoma, Hepatocellular ,Polymerase chain reaction ,Cells, Cultured ,beta Catenin ,Genetics ,Recombination, Genetic ,Mutation ,Liver Neoplasms ,MESH: APOBEC Deaminases ,Temperature ,MESH: CD4-Positive T-Lymphocytes ,APOBEC3 ,DNA, Neoplasm ,MESH: Temperature ,MESH: Hepatitis C, Chronic ,Nuclear DNA ,Neoplasm Proteins ,Oncology ,MESH: Recombination, Genetic ,MESH: Interferon-alpha ,MESH: Cytidine ,MESH: Cells, Cultured ,MESH: Mutation ,Carcinoma, Hepatocellular ,MESH: Hepatitis B, Chronic ,MESH: DNA, Neoplasm ,Context (language use) ,Genomics ,MESH: Nucleic Acid Denaturation ,Biology ,MESH: Phytohemagglutinins ,Hepatitis B, Chronic ,Cytidine Deaminase ,medicine ,Humans ,3DPCR ,Phytohemagglutinins ,MESH: Cytidine Deaminase ,cancer genomics ,MESH: Humans ,MESH: Cytosine Deaminase ,hypermutation ,Interferon-alpha ,MESH: Interleukin-2 ,MESH: Polymerase Chain Reaction ,Genetics and Genomics ,Hepatitis C, Chronic ,chemistry ,Interleukin-2 ,DNA - Abstract
International audience; Background: The revolution in cancer genomics shows that the dominant mutations are CG->TA transitions. The sources of these mutations are probably two host cell cytidine deaminases APOBEC3A and APOBEC3B. The former in particular can access nuclear DNA and monotonously introduce phenomenal numbers of C->T mutations in the signature 5'TpC context. These can be copied as G->A transitions in the 5'GpA context.Methods: DNA hypermutated by an APOBEC3 enzyme can be recovered by a technique called 3DPCR, which stands for differential DNA denaturation PCR. This method exploits the fact that APOBEC3-edited DNA is richer in A+T compared with the reference. We explore explicitly 3DPCR error using cloned DNA.Results: Here we show that the technique has a higher error rate compared with standard PCR and can generate DNA strands containing both C->T and G->A mutations in a 5'GpCpR context. Sequences with similar traits have been recovered from human tumour DNA using 3DPCR.Conclusions: Differential DNA denaturation PCR cannot be used to identify fixed C->T transitions in cancer genomes. Presently, the overall mutation frequency is ∼10(4)-10(5) base substitutions per cancer genome, or 0.003-0.03 kb(-1). By contrast, the 3DPCR error rate is of the order of 4-20 kb(-1) owing to constant selection for AT DNA and PCR-mediated recombination. Accordingly, sequences recovered by 3DPCR harbouring mixed C->T and G->A mutations associated with the 5'GpC represent artefacts.
- Published
- 2014
44. Final results of a phase I study evaluating INVAC-1, a novel DNA vaccine expressing an inactive form of human telomerase reverse transcriptase (hTERT) in patients with advanced solid tumors
- Author
-
Thierry Huet, Luis Augusto Teixeira, Mara Brizard, Jacques Medioni, Antoine Angelergues, Zineb Ghrieb, Jean-Jacques Kiladjian, Olivier Adotevi, Pierre Langlade Demoyen, Stéphane Culine, Marie-Agnès Dragon Durey, Julie Garibal, Rémy DeFrance, Ludovic Doucet, Valérie Doppler, Pierre Laurent-Puig, Caroline Laheurte, Stéphane Oudard, and Simon Wain-Hobson
- Subjects
Cancer Research ,Plasmid ,Oncology ,business.industry ,Cancer research ,Medicine ,Telomerase reverse transcriptase ,In patient ,business ,Tumor antigen ,DNA vaccination ,Phase i study - Abstract
2642 Background: INVAC-1 is an optimized DNA plasmid encoding an inactive form of human Telomerase Reverse Transcriptase (hTERT), a universal tumor antigen expressed in most of human tumors with little or no expression in somatic cells. We report here the final results of a First-In-Human Phase I study evaluating INVAC-1 as a single agent in patients (pts) with advanced solid tumors, ended in June 2018. Methods: A two center Phase I trial evaluated INVAC-1 given monthly for a minimum of 3 cycles and up to 9 cycles by intradermal injection followed by electroporation (n = 20) or using a needle-free injection system (n = 6). Primary objectives included safety, tolerability and dose limiting toxicities to identify the maximum tolerated dose and recommended phase 2 dose. Secondary objectives included immune response (assessed by IFN-γ Elispot) and anti-tumor activity. Immuno-monitoring included detection of autoantibodies, lymphocyte phenotyping and inflammatory cytokine levels in blood. Anti-tumor activity was evaluated through RECIST 1.1 adapted to immune response, and plasma circulating tumor DNA (ctDNA). Results: 26 pts with refractory/progressive tumors were enrolled and treated with 3 escalating doses of 100, 400 and 800 µg. 15 pts experienced stable disease according to RECIST. For 11 of them, the treatment was extended, up to 9 months. INVAC-1 was well tolerated with no dose-limiting toxicities. No significant biological signs of autoimmunity were observed. No significant modification in inflammatory plasma cytokines levels was observed after INVAC-1 administration. INVAC-1 triggered de novo or enhanced pre-existing CD4/CD8 specific anti-hTERT response in 63% of pts. This specific anti-hTERT immune response was enhanced ex vivo by adding the immune checkpoint inhibitor nivolumab. ctDNA was evaluated in 17 pts. We observed a ctDNA decrease in 6 cases, a stable level in 5 cases and an increase in 6 cases. Conclusions: Results indicate that INVAC-1 was well tolerated and immunogenic at the doses and schedule tested. Disease stabilization was obtained for the majority of pts (58%) according to RECIST criteria or ctDNA levels. Clinical trial information: NCT02301754.
- Published
- 2019
45. Orthologous Mammalian APOBEC3A Cytidine Deaminases Hypermutate Nuclear DNA
- Author
-
Simon Wain-Hobson, Jean-Pierre Vartanian, Vincent Caval, Rodolphe Suspène, Rétrovirologie Moléculaire, Institut Pasteur [Paris] (IP), and This work was supported by grants from the Institut Pasteur and Oséo (grant number FUI AAP 12). V.C. was supported by a postdoctoral grant from OSEO.
- Subjects
Models, Molecular ,Genome instability ,[SDV]Life Sciences [q-bio] ,MESH: DNA Breaks, Double-Stranded ,MESH: Protein Structure, Secondary ,MESH: Amino Acid Sequence ,Cytidine ,Protein Structure, Secondary ,Madin Darby Canine Kidney Cells ,MESH: Dogs ,chemistry.chemical_compound ,Gene cluster ,MESH: Animals ,MESH: Proteins ,DNA Breaks, Double-Stranded ,APOBEC3A ,MESH: Phylogeny ,Phylogeny ,Mammals ,Genetics ,double-strand breaks ,MESH: Genomic Instability ,MESH: DNA ,APOBEC3 ,Cytidine deaminase ,Nuclear DNA ,Deamination ,MESH: HEK293 Cells ,MESH: Cytidine ,MESH: Models, Molecular ,MESH: Mutation ,Molecular Sequence Data ,MESH: Sequence Alignment ,Biology ,MESH: Mammals ,Genomic Instability ,Dogs ,Cytidine Deaminase ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,MESH: Cytidine Deaminase ,MESH: Deamination ,MESH: Humans ,MESH: Molecular Sequence Data ,hypermutation ,nuclear DNA ,MESH: Madin Darby Canine Kidney Cells ,Proteins ,DNA ,methylcytidine ,genomic DNA ,HEK293 Cells ,chemistry ,MESH: HeLa Cells ,Mutation ,Sequence Alignment ,HeLa Cells - Abstract
International audience; The human APOBEC3 gene cluster locus encodes polynucleotide cytidine deaminases. Although many act as viral restriction factors through mutation of single-stranded DNA, recent reports have shown that human APOBEC3A was capable of efficiently hypermutating nuclear DNA and inducing DNA breaks in genomic DNA. In addition, the enzyme was unique in efficiently deaminating 5-methylcytidine in single-stranded DNA. To appreciate the evolutionary relevance of these activities, we analyzed A3A-related enzymes from the rhesus and tamarin monkey, horse, sheep, dog, and panda. All proved to be orthologous to the human enzyme in all these activities revealing strong conservation more than 148 My. Hence, their singular role in DNA catabolism is a well-established mechanism probably outweighing any deleterious or pathological roles such as genomic instability and cancer formation.
- Published
- 2013
46. PCR mediated recombination impacts the analysis of hepatitis B Virus covalently closed circular DNA
- Author
-
Valérie Thiers, Jean-Pierre Vartanian, Simon Wain-Hobson, Rodolphe Suspène, Rétrovirologie Moléculaire, Institut Pasteur [Paris] (IP), and This work was funded by the Institut Pasteur and the Centre National de Recherche Scientifique (CNRS).
- Subjects
0301 basic medicine ,PCR recombination ,Hepatitis B virus ,MESH: Hepatitis B, Chronic ,[SDV]Life Sciences [q-bio] ,030106 microbiology ,Short Report ,Biology ,medicine.disease_cause ,Real-Time Polymerase Chain Reaction ,Virus Replication ,Genome ,03 medical and health sciences ,chemistry.chemical_compound ,Hepatitis B, Chronic ,Transcription (biology) ,Virology ,medicine ,HBV ,Humans ,rcDNA ,MESH: Humans ,MESH: Hepatitis B ,MESH: Real-Time Polymerase Chain Reaction ,MESH: Virus Replication ,Virion ,cccDNA ,Hepatitis B ,medicine.disease ,Molecular biology ,MESH: DNA, Viral ,MESH: Hepatitis B virus ,030104 developmental biology ,Real-time polymerase chain reaction ,Infectious Diseases ,chemistry ,Viral replication ,DNA, Viral ,MESH: DNA, Circular ,MESH: Virion ,DNA, Circular ,DNA - Abstract
International audience; Background: The replication of HBV involves the production of covalently closed circular DNA (cccDNA) from the HBV genome through the repair of virion relaxed circular DNA (rcDNA) in the virion. As cccDNA is the transcription template for HBV genomes, it needs to be eliminated from hepatocytes if the eradication of chronic HBV infection is to be achieved. PCR quantitation of cccDNA copy number is the technique of choice for evaluating the efficiency of treatment regimens. The PCR target commonly used to identify cccDNA spans the gapped region of rcDNA and is considered to accurately distinguish between cccDNA and rcDNA. There is however, a potentially confounding issue in that PCR can generate larger targets from collections of small DNA fragments, a phenomenon known as PCR recombination.Results: The impact of PCR recombination towards the amplification of this cccDNA specific target was explored by mixing three marked, yet overlapping HBV DNA fragments. Thirteen of sixteen possible recombinants were identified by sequencing with frequencies ranging from 0.6 to 23%. To confirm this finding in vivo, HBV positive sera were treated with DNase I and submitted to quantitative real-time PCR. Under these conditions, it was possible to amplify the cccDNA specific segment without difficulty. As the virion contains uniquely rcDNA, amplification of the cccDNA target resulted from PCR recombination.Conclusions: PCR quantitation of cccDNA may be more difficult than hitherto thought. Current detection protocols need to be investigated so as to help in the management of chronic HBV infection.
- Published
- 2016
47. APOBEC3DE Antagonizes Hepatitis B Virus Restriction Factors APOBEC3F and APOBEC3G
- Author
-
Rodolphe Suspène, Vincent Caval, Pascal Pineau, Mohamed S. Bouzidi, Jean-Pierre Vartanian, Camille Hallez, Simon Wain-Hobson, Rétrovirologie Moléculaire, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Organisation Nucléaire et Oncogenèse / Nuclear Organization and Oncogenesis, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), This work was supported by grants from the Institut Pasteur and Centre National de la Recherche Scientifique (CNRS). M.S.B. and C.H. were supported by a bursary from la Ligue Nationale contre le Cancer (GB/MA/CD-11283) and Allocations de Recherche du Ministère de la Recherche, respectively., Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), and Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Hepatitis B virus ,viruses ,Somatic hypermutation ,APOBEC-3G Deaminase ,Biology ,medicine.disease_cause ,Virus Replication ,Cell Line ,Cytosine Deaminase ,03 medical and health sciences ,chemistry.chemical_compound ,Structural Biology ,Cytidine Deaminase ,medicine ,Animals ,Humans ,Molecular Biology ,Gene ,APOBEC3G ,ComputingMilieux_MISCELLANEOUS ,Genetics ,Gorilla gorilla ,030102 biochemistry & molecular biology ,Cytidine ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Cytidine deaminase ,Virology ,3. Good health ,HBcAg ,030104 developmental biology ,chemistry ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,DNA ,Protein Binding - Abstract
The APOBEC3 locus consists of seven genes (A3A-A3C, A3DE, A3F-A3H) that encode DNA cytidine deaminases. These enzymes deaminate single-stranded DNA, the result being DNA peppered with CG →TA mutations preferentially in the context of 5'TpC with the exception of APOBEC3G (A3G), which prefers 5'CpC dinucleotides. Hepatitis B virus (HBV) DNA is vulnerable to genetic editing by APOBEC3 cytidine deaminases, A3G being a major restriction factor. APOBEC3DE (A3DE) stands out in that it is catalytically inactive due to a fixed Tyr320Cys substitution in the C-terminal domain. As A3DE is closely related to A3F and A3G, which can form homo- and heterodimers and multimers, the impact of A3DE on HBV replication via modulation of other APOBEC3 restriction factors was investigated. A3DE binds to itself, A3F, and A3G and antagonizes A3F and, to a lesser extent, A3G restriction of HBV replication. A3DE suppresses A3F and A3G from HBV particles, leading to enhanced HBV replication. Ironically, while being part of a cluster of innate restriction factors, the A3DE phenotype is proviral. As the gorilla genome encodes the same Tyr320Cys substitution, this proviral phenotype seems to have been selected for.
- Published
- 2016
48. Structural Variability of the Herpes Simplex Virus 1 Genome In Vitro and In Vivo
- Author
-
Ayla Ergani, Aaron Bensimon, Sébastien Barradeau, Ahmed Azough, Fabrice Salvaire, Nicolas Alende, Charlotte Mahiet, Simon Wain-Hobson, Marc Labetoulle, Emmanuel Conseiller, and Nicolas Huot
- Subjects
Inverted repeat ,viruses ,Immunology ,Genome, Viral ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,Genome ,Cell Line ,Mice ,chemistry.chemical_compound ,Equivalent ,Virology ,medicine ,Animals ,Humans ,Polymorphism, Genetic ,Strain (chemistry) ,Nucleic Acid Hybridization ,Herpes Simplex ,Molecular biology ,In vitro ,Genome Replication and Regulation of Viral Gene Expression ,Disease Models, Animal ,Herpes simplex virus ,chemistry ,Insect Science ,DNA, Viral ,Homologous recombination ,DNA - Abstract
Herpes simplex virus 1 (HSV-1) is a human pathogen that leads to recurrent facial-oral lesions. Its 152-kb genome is organized in two covalently linked segments, each composed of a unique sequence flanked by inverted repeats. Replication of the HSV-1 genome produces concatemeric molecules in which homologous recombination events occur between the inverted repeats. This mechanism leads to four genome isomers (termed P, IS, IL, and ILS) that differ in the relative orientations of their unique fragments. Molecular combing analysis was performed on DNA extracted from viral particles and BSR, Vero, COS-7, and Neuro-2a cells infected with either strain SC16 or KOS of HSV-1, as well as from tissues of experimentally infected mice. Using fluorescence hybridization, isomers were repeatedly detected and distinguished and were accompanied by a large proportion of noncanonical forms (40%). In both cell and viral-particle extracts, the distributions of the four isomers were statistically equivalent, except for strain KOS grown in Vero and Neuro-2a cells, in which P and IS isomers were significantly overrepresented. In infected cell extracts, concatemeric molecules as long as 10 genome equivalents were detected, among which, strikingly, the isomer distributions were equivalent, suggesting that any such imbalance may occur during encapsidation. In vivo , for strain KOS-infected trigeminal ganglia, an unbalanced distribution distinct from the one in vitro was observed, along with a considerable proportion of noncanonical assortment.
- Published
- 2012
49. Clinical response and pharmacodynamic assessment of INVAC-1, a DNA plasmid encoding an inactive form of human telomerase reverse transcriptase (hTERT), on immune responses, immune tolerability, tumor burden and circulating tumor DNA (ctDNA) in patients with advanced solid tumors
- Author
-
B. Souttou, Caroline Laheurte, Pierre Langlade-Demoyen, Antoine Angelergues, Olivier Adotevi, R. Defrance, Stéphane Oudard, Marie-Agnès Dragon-Durey, T. Huet, Simon Wain-Hobson, M. Brizard, Stéphane Culine, L.O. Teixeira, V. Doppler, Jean-Jacques Kiladjian, Z. Ghrieb, Pierre Laurent-Puig, Ludovic Doucet, and J. Medioni
- Subjects
Telomerase ,business.industry ,Hematology ,chemistry.chemical_compound ,Plasmid ,Immune system ,Oncology ,Tolerability ,chemistry ,Pharmacodynamics ,Cancer research ,Medicine ,Telomerase reverse transcriptase ,A-DNA ,business ,DNA - Published
- 2018
50. APOBEC3G generates nonsense mutations in human T-cell leukemia virus type 1 proviral genomes in vivo
- Author
-
Kisato Nosaka, Jun Fan, Atsushi Koito, Masao Matsuoka, Yorifumi Satou, Jean-Pierre Vartanian, Junko Tanabe, Simon Wain-Hobson, and Guangyong Ma
- Subjects
viruses ,Genetic Vectors ,Molecular Sequence Data ,Immunology ,Nonsense mutation ,APOBEC-3G Deaminase ,Genome, Viral ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Proviruses ,Genes, Reporter ,immune system diseases ,Cytidine Deaminase ,Virology ,hemic and lymphatic diseases ,medicine ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Gene ,APOBEC3G ,030304 developmental biology ,Genetics ,Human T-lymphotropic virus 1 ,0303 health sciences ,Mutation ,Base Sequence ,biology ,Genetic Variation ,Provirus ,biology.organism_classification ,HTLV-I Infections ,Virus-Cell Interactions ,3. Good health ,Mutagenesis ,030220 oncology & carcinogenesis ,Insect Science ,Carcinogenesis - Abstract
Human T-cell leukemia virus type 1 (HTLV-1) induces cell proliferation after infection, leading to efficient transmission by cell-to-cell contact. After a long latent period, a fraction of carriers develop adult T-cell leukemia (ATL). Genetic changes in the tax gene in ATL cells were reported in about 10% of ATL cases. To determine genetic changes that may occur throughout the provirus, we determined the entire sequence of the HTLV-1 provirus in 60 ATL cases. Abortive genetic changes, including deletions, insertions, and nonsense mutations, were frequent in all viral genes except the HBZ gene, which is transcribed from the minus strand of the virus. G-to-A base substitutions were the most frequent mutations in ATL cells. The sequence context of G-to-A mutations was in accordance with the preferred target sequence of human APOBEC3G (hA3G). The target sequences of hA3G were less frequent in the plus strand of the HBZ coding region than in other coding regions of the HTLV-1 provirus. Nonsense mutations in viral genes including tax were also observed in proviruses from asymptomatic carriers, indicating that these mutations were generated during reverse transcription and prior to oncogenesis. The fact that hA3G targets the minus strand during reverse transcription explains why the HBZ gene is not susceptible to such nonsense mutations. HTLV-1-infected cells likely take advantage of hA3G to escape from the host immune system by losing expression of viral proteins.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.