1
|
Wang TT, Hirons A, Doerflinger M, Morris KV, Ledger S, Purcell DFJ, Kelleher AD, Ahlenstiel CL. Current State of Therapeutics for HTLV-1. Viruses 2024; 16:1616. [PMID: 39459949 PMCID: PMC11512412 DOI: 10.3390/v16101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Human T cell leukaemia virus type-1 (HTLV-1) is an oncogenic retrovirus that causes lifelong infection in ~5-10 million individuals globally. It is endemic to certain First Nations populations of Northern and Central Australia, Japan, South and Central America, Africa, and the Caribbean region. HTLV-1 preferentially infects CD4+ T cells and remains in a state of reduced transcription, often being asymptomatic in the beginning of infection, with symptoms developing later in life. HTLV-1 infection is implicated in the development of adult T cell leukaemia/lymphoma (ATL) and HTLV-1-associated myelopathies (HAM), amongst other immune-related disorders. With no preventive or curative interventions, infected individuals have limited treatment options, most of which manage symptoms. The clinical burden and lack of treatment options directs the need for alternative treatment strategies for HTLV-1 infection. Recent advances have been made in the development of RNA-based antiviral therapeutics for Human Immunodeficiency Virus Type-1 (HIV-1), an analogous retrovirus that shares modes of transmission with HTLV-1. This review highlights past and ongoing efforts in the development of HTLV-1 therapeutics and vaccines, with a focus on the potential for gene therapy as a new treatment modality in light of its successes in HIV-1, as well as animal models that may help the advancement of novel antiviral and anticancer interventions.
Collapse
Affiliation(s)
- Tiana T. Wang
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Ashley Hirons
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Melbourne, VIC 3050, Australia
| | - Kevin V. Morris
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia;
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Damian F. J. Purcell
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Anthony D. Kelleher
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Chantelle L. Ahlenstiel
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
2
|
Plant E, Bellefroid M, Van Lint C. A complex network of transcription factors and epigenetic regulators involved in bovine leukemia virus transcriptional regulation. Retrovirology 2023; 20:11. [PMID: 37268923 PMCID: PMC10236774 DOI: 10.1186/s12977-023-00623-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 05/09/2023] [Indexed: 06/04/2023] Open
Abstract
Bovine Leukemia Virus (BLV) is the etiological agent of enzootic bovine leukosis, a disease characterized by the neoplastic proliferation of B cells in cattle. While most European countries have introduced efficient eradication programs, BLV is still present worldwide and no treatment is available. A major feature of BLV infection is the viral latency, which enables the escape from the host immune system, the maintenance of a persistent infection and ultimately the tumoral development. BLV latency is a multifactorial phenomenon resulting in the silencing of viral genes due to genetic and epigenetic repressions of the viral promoter located in the 5' Long Terminal Repeat (5'LTR). However, viral miRNAs and antisense transcripts are expressed from two different proviral regions, respectively the miRNA cluster and the 3'LTR. These latter transcripts are expressed despite the viral latency affecting the 5'LTR and are increasingly considered to take part in tumoral development. In the present review, we provide a summary of the experimental evidence that has enabled to characterize the molecular mechanisms regulating each of the three BLV transcriptional units, either through cis-regulatory elements or through epigenetic modifications. Additionally, we describe the recently identified BLV miRNAs and antisense transcripts and their implications in BLV-induced tumorigenesis. Finally, we discuss the relevance of BLV as an experimental model for the closely related human T-lymphotropic virus HTLV-1.
Collapse
Affiliation(s)
- Estelle Plant
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041, Gosselies, Belgium
| | - Maxime Bellefroid
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041, Gosselies, Belgium
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041, Gosselies, Belgium.
| |
Collapse
|
3
|
Gomes YCP, Silva MTT, Leite ACCB, Lima MASD, Araújo AQC, Silva Filho IL, Vicente ACP, Espíndola ODM. Polymorphisms in HTLV-1 Tax-responsive elements in HTLV-1-associated myelopathy/tropical spastic paraparesis patients are associated with reduced proviral load but not with disease progression. J Gen Virol 2021; 102. [PMID: 34494950 DOI: 10.1099/jgv.0.001649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) provirus expression is mainly directed by Tax-responsive elements (TRE) within the long terminal repeats (LTR). Mutations in TRE can reduce provirus expression and since a high proviral load (PVL) is a risk factor for the development of HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), we evaluated polymorphisms in the 5' LTR and the association with PVL and disease progression. HTLV-1 LTR and tax sequences derived from asymptomatic carriers (AC) and HAM/TSP patients followed in a longitudinal study were analysed according to PVL and clinical severity. Individuals infected with HTLV-1 presenting the canonical TRE, considering strain ATK-1 as the consensus, displayed sustained higher PVL. By contrast, an LTR A125G mutation in TRE was associated with slightly reduced PVL only in HAM/TSP patients, although it did not influence the speed of disease progression. Moreover, this polymorphism was frequent in Latin American strains of the HTLV-1 Cosmopolitan Transcontinental subtype. Therefore, polymorphisms in the 5' TRE of HTLV-1 may represent one of the factors influencing PVL in HAM/TSP patients, especially in the Latin American population. Indeed, higher PVL in the peripheral blood has been associated with an increased inflammatory activity in the spinal cord and to a poorer prognosis in HAM/TSP. However, this event was not associated with TRE polymorphisms.
Collapse
Affiliation(s)
- Yago Côrtes Pinheiro Gomes
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Marcus Tulius Teixeira Silva
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Ana Claudia Celestino Bezerra Leite
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Marco Antonio Sales Dantas Lima
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Abelardo Queiroz Campos Araújo
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Isaac Lima Silva Filho
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Ana Carolina Paulo Vicente
- Laboratory of Molecular Genetics of Microorganisms, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| | - Otávio de Melo Espíndola
- Laboratory for Clinical Research in Neuroinfections, Evandro Chagas National Institute of Infectious Diseases (INI), Oswaldo Cruz Foundation (FIOCRUZ), Brazil
| |
Collapse
|
4
|
Analysis of Nucleotide Sequence of Tax, miRNA and LTR of Bovine Leukemia Virus in Cattle with Different Levels of Persistent Lymphocytosis in Russia. Pathogens 2021; 10:pathogens10020246. [PMID: 33672613 PMCID: PMC7924208 DOI: 10.3390/pathogens10020246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 11/17/2022] Open
Abstract
Bovine Leukemia Virus (BLV) is the etiological agent of enzootic bovine leucosis (EBL), a lymphoproliferative disease of the bovine species. In BLV-infected cells, the long terminal repeat (LTR), the viral Tax protein and viral miRNAs promote viral and cell proliferation as well as tumorigenesis. Although their respective roles are decisive in BLV biology, little is known about the genetic sequence variation of these parts of the BLV genome and their impact on disease outcome. Therefore, the objective of this study was to assess the relationship between disease progression and sequence variation of the BLV Tax, miRNA and LTR regions in infected animals displaying either low or high levels of persistent lymphocytosis (PL). A statistically significant association was observed between the A(+187)C polymorphism in the downstream activator sequence (DAS) region in LTR (p-value = 0.00737) and high lymphocytosis. Our study also showed that the mutation A(−4)G in the CAP site occurred in 70% of isolates with low PL and was not found in the high PL group. Conversely, the mutations G(−133)A/C in CRE2 (46.7%), C(+160)T in DAS (30%) and A(310)del in BLV-mir-B4-5p, A(357)G in BLV-mir-B4-3p, A(462)G in BLV-mir-B5-5p, and GA(497–498)AG in BLV-mir-B5-3p (26.5%) were often seen in isolates with high PL and did not occur in the low PL group. In conclusion, we found several significant polymorphisms among BLV genomic sequences in Russia that would explain a progression towards higher or lower lymphoproliferation. The data presented in this article enabled the classification between two different genotypes; however, clear association between genotypes and the PL development was not found.
Collapse
|
5
|
Blazhko N, Shatokhin K, Khripko Y, Ngirande C, Kochnev N. Mutational and phylogenetic status of west siberian strains of BLV. BIO WEB OF CONFERENCES 2021. [DOI: 10.1051/bioconf/20213606025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The study is devoted of full-genome BLV sequences circulating in cattle populations of the Novosibirsk region, Russia. The phylogenetic tree shows that the West Siberian isolates are quite closely related to such previously isolated strains as AF399704 (Brazil), AP018007, AP018016, AP018019, LC007988, LC007991 (Japan) and EF065638 (Belgium) we calculations show that the number of mutations that could independently occur in parallel evolving BLV strains significantly exceeds the expected number based on the probability of corresponding substitutions. It was also found that the studied isolates have some mutations, the presence of which, at first glance, is possible only with their divergent development in different independently evolving branches. However, calculations show that the probability of an independent origin of an identical mutation is extremely small, which indicates the possibility of exchanging RNA sites between isolates circulating in West Siberian cattle populations.
Collapse
|
6
|
Pluta A, Willems L, Douville RN, Kuźmak J. Effects of Naturally Occurring Mutations in Bovine Leukemia Virus 5'-LTR and Tax Gene on Viral Transcriptional Activity. Pathogens 2020; 9:pathogens9100836. [PMID: 33066207 PMCID: PMC7656303 DOI: 10.3390/pathogens9100836] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 01/28/2023] Open
Abstract
Bovine leukemia virus (BLV) is a deltaretrovirus infecting bovine B cells and causing enzootic bovine leucosis (EBL). The long terminal repeat (LTR) plays an indispensable role in viral gene expression. The BLV Tax protein acts as the main transactivator of LTR-driven transcription of BLV viral genes. The aim of this study was to analyze mutations in the BLV LTR region and tax gene to determine their association with transcriptional activity. LTRs were obtained from one hundred and six BLV isolates and analyzed for their genetic variability. Fifteen variants were selected and characterized based on mutations in LTR regulatory elements, and further used for in vitro transcription assays. Reporter vectors containing the luciferase gene under the control of each variant BLV promoter sequence, in addition to variant Tax expression vectors, were constructed. Both types of plasmids were used for cotransfection of HeLa cells and the level of luciferase activity was measured as a proxy of transcriptional activity. Marked differences in LTR promoter activity and Tax transactivation activity were observed amongst BLV variants. These results demonstrate that mutations in both the BLV LTR and tax gene can affect the promoter activity, which may have important consequences on proviral load, viral fitness, and transmissibility in BLV-infected cattle.
Collapse
Affiliation(s)
- Aneta Pluta
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Puławy, Poland;
- Correspondence:
| | - Luc Willems
- Molecular and Cellular Epigenetics (Interdisciplinary Cluster for Applied Genoproteomics, GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000 Liege, Belgium;
| | - Renée N. Douville
- Department of Biology, The University of Winnipeg, Winnipeg, MB R3B 2E9, Canada;
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jacek Kuźmak
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Puławy, Poland;
| |
Collapse
|
7
|
Regulation of Expression and Latency in BLV and HTLV. Viruses 2020; 12:v12101079. [PMID: 32992917 PMCID: PMC7601775 DOI: 10.3390/v12101079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Human T-lymphotrophic virus type 1 (HTLV-1) and Bovine leukemia virus (BLV) belong to the Deltaretrovirus genus. HTLV-1 is the etiologic agent of the highly aggressive and currently incurable cancer adult T-cell leukemia (ATL) and a neurological disease HTLV-1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP). BLV causes neoplastic proliferation of B cells in cattle: enzootic bovine leucosis (EBL). Despite the severity of these conditions, infection by HTLV-1 and BLV appear in most cases clinically asymptomatic. These viruses can undergo latency in their hosts. The silencing of proviral gene expression and maintenance of latency are central for the establishment of persistent infection, as well as for pathogenesis in vivo. In this review, we will present the mechanisms that control proviral activation and retroviral latency in deltaretroviruses, in comparison with other exogenous retroviruses. The 5′ long terminal repeats (5′-LTRs) play a main role in controlling viral gene expression. While the regulation of transcription initiation is a major mechanism of silencing, we discuss topics that include (i) the epigenetic control of the provirus, (ii) the cis-elements present in the LTR, (iii) enhancers with cell-type specific regulatory functions, (iv) the role of virally-encoded transactivator proteins, (v) the role of repressors in transcription and silencing, (vi) the effect of hormonal signaling, (vii) implications of LTR variability on transcription and latency, and (viii) the regulatory role of non-coding RNAs. Finally, we discuss how a better understanding of these mechanisms may allow for the development of more effective treatments against Deltaretroviruses.
Collapse
|
8
|
Ablation of non-coding RNAs affects bovine leukemia virus B lymphocyte proliferation and abrogates oncogenesis. PLoS Pathog 2020; 16:e1008502. [PMID: 32407379 PMCID: PMC7252678 DOI: 10.1371/journal.ppat.1008502] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/27/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022] Open
Abstract
Viruses have developed different strategies to escape from immune response. Among these, viral non-coding RNAs are invisible to the immune system and may affect the fate of the host cell. Bovine leukemia virus (BLV) encodes both short (miRNAs) and long (antisense AS1 and AS2) non-coding RNAs. To elucidate the mechanisms associated with BLV non-coding RNAs, we performed phenotypic and transcriptomic analyzes in a reverse genetics system. RNA sequencing of B-lymphocytes revealed that cell proliferation is the most significant mechanism associated with ablation of the viral non-coding RNAs. To assess the biological relevance of this observation, we determined the cell kinetic parameters in vivo using intravenous injection of BrdU and CFSE. Fitting the data to a mathematical model provided the rates of cell proliferation and death. Our data show that deletion of miRNAs correlates with reduced proliferation of the infected cell and lack of pathogenesis. BLV is a retrovirus that integrates into the genomic DNA of B-lymphocytes from a series of ruminant species (cattle, sheep, zebu, water buffalo and yack). Expression of viral proteins is almost undetectable in infected animals. In contrast, the BLV genome contains a cluster of 10 microRNAs that are abundantly transcribed in BLV-infected cells in vivo. In this report, we show that these microRNAs primarily regulate host cell proliferation. Ablation of the viral microRNAs affects BLV replication and suppresses leukemia development.
Collapse
|
9
|
Abdala A, Alvarez I, Brossel H, Calvinho L, Carignano H, Franco L, Gazon H, Gillissen C, Hamaidia M, Hoyos C, Jacques JR, Joris T, Laval F, Petersen M, Porquet F, Porta N, Ruiz V, Safari R, Suárez Archilla G, Trono K, Willems L. BLV: lessons on vaccine development. Retrovirology 2019; 16:26. [PMID: 31590667 PMCID: PMC6781361 DOI: 10.1186/s12977-019-0488-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/10/2019] [Indexed: 01/12/2023] Open
Abstract
Vaccination against retroviruses is a challenge because of their ability to stably integrate into the host genome, undergo long-term latency in a proportion of infected cells and thereby escape immune response. Since clearance of the virus is almost impossible once infection is established, the primary goal is to achieve sterilizing immunity. Besides efficacy, safety is the major issue since vaccination has been associated with increased infection or reversion to pathogenicity. In this review, we discuss the different issues that we faced during the development of an efficient vaccine against bovine leukemia virus (BLV). We summarize the historical failures of inactivated vaccines, the efficacy and safety of a live-attenuated vaccine and the economical constraints of further industrial development.
Collapse
Affiliation(s)
- Alejandro Abdala
- Estacion Experimental Agropecuaria Rafaela, INTA, 2300, Rafaela, Argentina
| | - Irene Alvarez
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Hélène Brossel
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Luis Calvinho
- Estacion Experimental Agropecuaria Rafaela, INTA, 2300, Rafaela, Argentina
| | - Hugo Carignano
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Lautaro Franco
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Hélène Gazon
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Christelle Gillissen
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Malik Hamaidia
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Clotilde Hoyos
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Jean-Rock Jacques
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Thomas Joris
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Florent Laval
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Marcos Petersen
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Florent Porquet
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | - Natalia Porta
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Vanesa Ruiz
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Roghaiyeh Safari
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium
| | | | - Karina Trono
- Instituto de Virología e Innovaciones tecnológicas, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA-CONICET, C.C. 1712, Castelar, Argentina
| | - Luc Willems
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liège (ULiège), 4000, Liege, Belgium. .,Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA) of University of Liège (ULiège), B34, 1 avenue de l'Hôpital, Sart-Tilman, 4000, Liege, Belgium.
| |
Collapse
|
10
|
Sato H, Watanuki S, Bai L, Borjigin L, Ishizaki H, Matsumoto Y, Hachiya Y, Sentsui H, Aida Y. A sensitive luminescence syncytium induction assay (LuSIA) based on a reporter plasmid containing a mutation in the glucocorticoid response element in the long terminal repeat U3 region of bovine leukemia virus. Virol J 2019; 16:66. [PMID: 31109347 PMCID: PMC6528319 DOI: 10.1186/s12985-019-1172-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/30/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bovine leukemia virus (BLV) causes enzootic bovine leukosis, the most common neoplastic disease of cattle. Previously, we reported the luminescence syncytium induction assay (LuSIA), an assay for BLV infectivity based on CC81-BLU3G cells, which form syncytia expressing enhanced green fluorescent protein (EGFP) when co-cultured with BLV-infected cells. To develop a more sensitive LuSIA, we here focused on the glucocorticoid response element (GRE) within the U3 region of the BLV long terminal repeat (LTR). METHODS We changed five nucleotide sites of the GRE in a pBLU3-EGFP reporter plasmid containing the BLV-LTR U3 region promoter by site-directed mutagenesis and we then constructed a new reporter plasmid (pBLU3GREM-EGFP) in which the EGFP reporter gene was expressed under control of the GRE-mutated LTR-U3 promoter. We also established a new CC81-derived reporter cell line harboring the GRE-mutated LTR-U3 promoter (CC81-GREMG). To evaluate the sensibility, the utility and the specificity of the LuSIA using CC81-GREMG, we co-cultured CC81-GREMG cells with BLV-persistently infected cells, free-viruses, white blood cells (WBCs) from BLV-infected cows, and bovine immunodeficiency-like virus (BIV)- and bovine foamy virus (BFV)-infected cells. RESULTS We successfully constructed a new reporter plasmid harboring a mutation in the GRE and established a new reporter cell line, CC81-GREMG; this line was stably transfected with pBLU3GREM-EGFP in which the EGFP gene is expressed under control of the GRE-mutated LTR-U3 promoter and enabled direct visualization of BLV infectivity. The new LuSIA protocol using CC81-GREMG cells measures cell-to-cell infectivity and cell-free infectivity of BLV more sensitively than previous protocol using CC81-BLU3G. Furthermore, it did not respond to BIV and BFV infections, indicating that the LuSIA based on CC81-GREMG is specific for BLV infectivity. Moreover, we confirmed the utility of a new LuSIA based on CC81-GREMG cells using white blood cells (WBCs) from BLV-infected cows. Finally, the assay was useful for assessing the activity of neutralizing antibodies in plasma collected from BLV-infected cows. CONCLUSION The new LuSIA protocol is quantitative and more sensitive than the previous assay based on CC81-BLU3G cells and should facilitate development of several new BLV assays.
Collapse
Affiliation(s)
- Hirotaka Sato
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan.,Virus Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan
| | - Sonoko Watanuki
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan.,Virus Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan.,Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Lanlan Bai
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan.,Virus Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan
| | - Liushiqi Borjigin
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan
| | - Hiroshi Ishizaki
- Grazing Animal Unit, Division of Grassland Farming, Institute of Livestock and Grassland Science, NARO, 768 Senbonmatsu, Nasushiobara, Tochigi, 329-2793, Japan
| | - Yasunobu Matsumoto
- Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Yuma Hachiya
- Department of Veterinary Medicine, Nihon University, Kameino 1866, Fujisawa, Kanagawa, 252-0880, Japan
| | - Hiroshi Sentsui
- Department of Veterinary Medicine, Nihon University, Kameino 1866, Fujisawa, Kanagawa, 252-0880, Japan
| | - Yoko Aida
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan. .,Virus Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 3510198, Japan. .,Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
11
|
Blazhko NV, Vyshegurov SK, Donchenko AS, Shatokhin KS, Krytsyna TI, Ryabinina VA. Association of haplotypes for SNPs in the LTR regions of bovine leukemia virus with hematological indices of cattle. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Molecular typing of BLV samples isolated from Holsteinized Russian Black Pied cattle was carried out, and various cytofluorometric and morphological blood indices were examined. We performed the total count of white blood cells (WBC), lymphocyte (lymf), granulocyte (gran), monocyte (mon), red blood cell (RBC), hemoglobin (HGB), hematocrit (HTC), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), red blood cell distribution width (RDW), platelet count (PLT), mean platelet volume (MPV), platelet distribution width (PDW), and platelet crit count (PCT). The LTR-region of BLV was haplotyped. Only viruses of haplotypes I (0.33±0.03) and III (0.67±0.03) of the eight possible were detected. The ratio of hematologically sick, healthy, and suspected carriers of BLV of haplotypes I and II was comparable with the results of other researchers. The numbers of leukocytes, erythrocytes and platelets in the blood of carriers of haplotype III exceeded the corresponding parameters of cattle affected by the virus of haplotype I. It is interesting to note that the difference in the hemolytic status of animals was manifested not only by the concentration of leukocytes as direct immune agents but also by the count of erythrocytes and platelets, which are not directly involved in the immune response. The number of particles of haplotype III of the BLV circulating in the blood of infected individuals exceeded that of the carriers of haplotype I. In this connection, an assumption was made about the evolutionary advantage of the more virulent haplotype III. However, the results of our own research in conjunction with the data of other scientists indicate that the high virulence of individual virus strains is a consequence of the tendency to implement the maximum possible intensity of the synthesis of virus particles but not of the high damaging effect alone. It is shown that high lethality is evolutionarily disadvantageous for viruses, since the extinction of the carrier as a biological species is fraught with the disappearance of the virus itself.
Collapse
Affiliation(s)
| | | | - A. S. Donchenko
- Novosibirsk State Agrarian University; Siberian Federal Research Centre for AgroBiotechnology, RAS
| | | | | | | |
Collapse
|
12
|
Pluta A, Rola-Łuszczak M, Douville RN, Kuźmak J. Bovine leukemia virus long terminal repeat variability: identification of single nucleotide polymorphisms in regulatory sequences. Virol J 2018; 15:165. [PMID: 30359262 PMCID: PMC6202831 DOI: 10.1186/s12985-018-1062-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/25/2018] [Indexed: 12/03/2022] Open
Abstract
Background Limited data are available on the incidence of variations in nucleotide sequences of long terminal repeat (LTR) regions of Bovine Leukemia Virus (BLV). Consequently, the possible impact of SNPs on BLV LTR function are poorly elucidated. Thus, a detailed and representative study of full-length LTR sequences obtained from sixty-four BLV isolates from different geographical regions of Poland, Moldova, Croatia, Ukraine and Russia were analyzed for their genetic variability. Methods Overlap extension PCR, sequencing and Bayesian phylogenetic reconstruction of LTR sequences were performed. These analyses were followed by detailed sequence comparison, estimation of genetic heterogeneity and identification of transcription factor binding site (TFBS) modifications. Results Phylogenetic analysis of curated LTR sequences and those available in the GenBank database reflected the acknowledged env gene classification of BLV into 10 genotypes, and further clustered analysed sequences into three genotypes - G4, G7 and G8. Additional molecular studies revealed the presence of 97 point mutations distributed at 89 positions throughout all 64 LTR sequences. The highest rate of variability was noted in U3 and U5 subregions. However, the variability in regulatory sequences (VR) was assessed as lower than the variability within non-regulatory sequences (VNR) for both, U3 and U5 subregions. In contrast, VR value for R subregion, as well as for the total LTR, was higher than the VNR suggesting the existence of positive selection. Twelve unique SNPs for these LTR sequences localized in regulatory and non-regulatory elements were identified. The presence of different types of substitutions lead to the abrogation of present or to the creation of additional TFBS. Conclusion This study represents the largest study of LTR genetic variability of BLV field isolates from Eastern part of Europe. Phylogenetic analysis of LTRs supports the clustering BLV variants based on their geographic origin. The SNP screening showed variations modifying LTR regulatory sequences, as well as altering TFBS. These features warrant further exploration as they could be related to proviral load and distinctive regulation of BLV transcription and replication. Electronic supplementary material The online version of this article (10.1186/s12985-018-1062-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aneta Pluta
- Department of Biochemistry, National Veterinary Research Institute, Puławy, Poland.
| | | | - Renée N Douville
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jacek Kuźmak
- Department of Biochemistry, National Veterinary Research Institute, Puławy, Poland
| |
Collapse
|
13
|
Polat M, Takeshima SN, Hosomichi K, Kim J, Miyasaka T, Yamada K, Arainga M, Murakami T, Matsumoto Y, de la Barra Diaz V, Panei CJ, González ET, Kanemaki M, Onuma M, Giovambattista G, Aida Y. A new genotype of bovine leukemia virus in South America identified by NGS-based whole genome sequencing and molecular evolutionary genetic analysis. Retrovirology 2016; 13:4. [PMID: 26754835 PMCID: PMC4709907 DOI: 10.1186/s12977-016-0239-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/05/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bovine leukemia virus (BLV) is a member of retroviridae family, together with human T cell leukemia virus types 1 and 2 (HTLV-1 and -2) belonging to the genes deltaretrovirus, and infects cattle worldwide. Previous studies have classified the env sequences of BLV provirus from different geographic locations into eight genetic groups. To investigate the genetic variability of BLV in South America, we performed phylogenetic analyses of whole genome and partial env gp51 sequences of BLV strains isolated from Peru, Paraguay and Bolivia, for which no the molecular characteristics of BLV have previously been published, and discovered a novel BLV genotype, genotype-9, in Bolivia. RESULTS In Peru and Paraguay, 42.3 % (139/328) and over 50 % (76/139) of samples, respectively, were BLV positive. In Bolivia, the BLV infection rate was up to 30 % (156/507) at the individual level. In Argentina, 325/420 samples were BLV positive, with a BLV prevalence of 77.4 % at the individual level and up to 90.9 % at herd level. By contrast, relatively few BLV positive samples were detected in Chile, with a maximum of 29.1 % BLV infection at the individual level. We performed phylogenetic analyses using two different approaches, maximum likelihood (ML) tree and Bayesian inference, using 35 distinct partial env gp51 sequences from BLV strains isolated from Peru, Paraguay, and Bolivia, and 74 known BLV strains, representing eight different BLV genotypes from various geographical locations worldwide. The results indicated that Peruvian and Paraguayan BLV strains were grouped into genotypes-1, -2, and -6, while those from Bolivia were clustered into genotypes-1, -2, and -6, and a new genotype, genotype-9. Interestingly, these results were confirmed using ML phylogenetic analysis of whole genome sequences obtained by next generation sequencing of 25 BLV strains, assigned to four different genotypes (genotypes-1, -2, -6, and -9) from Peru, Paraguay, and Bolivia. Comparative analyses of complete genome sequences clearly showed some specific substitutions, in both structural and non-structural BLV genes, distinguishing the novel genotype-9 from known genotypes. CONCLUSIONS Our results demonstrate widespread BLV infection in South American cattle and the existence of a new BLV genotype-9 in Bolivia. We conclude that at least seven BLV genotypes (genotypes-1, -2, -4, -5, -6, -7, and -9) are circulating in South America.
Collapse
Affiliation(s)
- Meripet Polat
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan. .,Laboratory of Viral Infectious Diseases, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Wako, Saitama, 351-0198, Japan.
| | - Shin-Nosuke Takeshima
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan. .,Laboratory of Viral Infectious Diseases, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Wako, Saitama, 351-0198, Japan.
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8640, Japan.
| | - Jiyun Kim
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Taku Miyasaka
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Kazunori Yamada
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Mariluz Arainga
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Tomoyuki Murakami
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Yuki Matsumoto
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | | | - Carlos Javier Panei
- Department of Virology, Faculty of Veterinary Sciences, National University of La Plata, 60 and 118, CC 296, 1900, La Plata, Argentina. .,IGEVET, CCT La Plata-CONICET, Facultad de Ciencias Veterinarias, National University of La Plata, 60 and 118, CC 296, 1900, La Plata, Argentina.
| | - Ester Teresa González
- Department of Virology, Faculty of Veterinary Sciences, National University of La Plata, 60 and 118, CC 296, 1900, La Plata, Argentina.
| | - Misao Kanemaki
- Institute for Animal Science, Shitara-cho, Aichi, 441-2433, Japan.
| | - Misao Onuma
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Guillermo Giovambattista
- IGEVET, CCT La Plata-CONICET, Facultad de Ciencias Veterinarias, National University of La Plata, 60 and 118, CC 296, 1900, La Plata, Argentina.
| | - Yoko Aida
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan. .,Laboratory of Viral Infectious Diseases, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
14
|
Abstract
Different animal models have been proposed to investigate the mechanisms of Human T-lymphotropic Virus (HTLV)-induced pathogenesis: rats, transgenic and NOD-SCID/γcnull (NOG) mice, rabbits, squirrel monkeys, baboons and macaques. These systems indeed provide useful information but have intrinsic limitations such as lack of disease relevance, species specificity or inadequate immune response. Another strategy based on a comparative virology approach is to characterize a related pathogen and to speculate on possible shared mechanisms. In this perspective, bovine leukemia virus (BLV), another member of the deltaretrovirus genus, is evolutionary related to HTLV-1. BLV induces lymphoproliferative disorders in ruminants providing useful information on the mechanisms of viral persistence, genetic determinants of pathogenesis and potential novel therapies.
Collapse
|
15
|
Mutation of a Single Envelope N-Linked Glycosylation Site Enhances the Pathogenicity of Bovine Leukemia Virus. J Virol 2015; 89:8945-56. [PMID: 26085161 DOI: 10.1128/jvi.00261-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/09/2015] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Viruses have coevolved with their host to ensure efficient replication and transmission without inducing excessive pathogenicity that would indirectly impair their persistence. This is exemplified by the bovine leukemia virus (BLV) system in which lymphoproliferative disorders develop in ruminants after latency periods of several years. In principle, the equilibrium reached between the virus and its host could be disrupted by emergence of more pathogenic strains. Intriguingly but fortunately, such a hyperpathogenic BLV strain was never observed in the field or designed in vitro. In this study, we sought to understand the role of envelope N-linked glycosylation with the hypothesis that this posttranslational modification could either favor BLV infection by allowing viral entry or allow immune escape by using glycans as a shield. Using reverse genetics of an infectious molecular provirus, we identified a N-linked envelope glycosylation site (N230) that limits viral replication and pathogenicity. Indeed, mutation N230E unexpectedly leads to enhanced fusogenicity and protein stability. IMPORTANCE Infection by retroviruses requires the interaction of the viral envelope protein (SU) with a membrane-associated receptor allowing fusion and release of the viral genomic RNA into the cell. We show that N-linked glycosylation of the bovine leukemia virus (BLV) SU protein is, as expected, essential for cell infection in vitro. Consistently, mutation of all glycosylation sites of a BLV provirus destroys infectivity in vivo. However, single mutations do not significantly modify replication in vivo. Instead, a particular mutation at SU codon 230 increases replication and accelerates pathogenesis. This unexpected observation has important consequences in terms of disease control and managing.
Collapse
|
16
|
Bovine leukemia virus: a major silent threat to proper immune responses in cattle. Vet Immunol Immunopathol 2014; 163:103-14. [PMID: 25554478 DOI: 10.1016/j.vetimm.2014.11.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/27/2014] [Accepted: 11/26/2014] [Indexed: 11/22/2022]
Abstract
Bovine leukemia virus (BLV) infection is widespread in the US dairy industry and the majority of producers do not actively try to manage or reduce BLV incidence within their herds. However, BLV is estimated to cost the dairy industry hundreds of millions of dollars annually and this is likely a conservative estimate. BLV is not thought to cause animal distress or serious pathology unless infection progresses to leukemia or lymphoma. However, a wealth of research supports the notion that BLV infection causes widespread abnormal immune function. BLV infection can impact cells of both the innate and adaptive immune system and alter proper functioning of uninfected cells. Despite strong evidence of abnormal immune signaling and functioning, little research has investigated the large-scale effects of BLV infection on host immunity and resistance to other infectious diseases. This review focuses on mechanisms of immune suppression associated with BLV infection, specifically aberrant signaling, proliferation and apoptosis, and the implications of switching from BLV latency to activation. In addition, this review will highlight underdeveloped areas of research relating to BLV infection and how it causes immune suppression.
Collapse
|
17
|
Barreto FK, Rego FF, Fonseca LM, Galvão-Castro-Filho B, Araújo TH, Mota-Miranda ACA, Monteiro-Cunha JP, Alcantara LCJ. Molecular characterization of the human T cell lymphotropic virus type 2 long terminal repeat region: A discussion about possible influences at viral gene expression. AIDS Res Hum Retroviruses 2014; 30:92-6. [PMID: 23808326 DOI: 10.1089/aid.2013.0181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
This study aimed to identify nucleotide signatures in the promoter region of human T cell lymphotropic virus type 2 (HTLV-2) isolated from infected individuals from Salvador, Brazil and in sequences from the GenBank database. DNA samples from HTLV-2-infected individuals were submitted to nested polymerase chain reaction (PCR) and sequencing, and molecular analyses were performed using bioinformatics tools. The phylogeny of HTLV-2 strains isolated from patients from Salvador reveals that all sequences were subtype c. One hundred and fifty-one sequences from GenBank were selected, among which 30 belong to subtype a, 88 to subtype b, 32 to subtype c, and one to subtype d. Subtype-specific signatures were identified as well as mutations resulting in loss or gain of motifs important to transcription regulation. The subtypes a and b have two E box motifs, while subtypes c and d have only one. These polymorphisms may impact viral fitness and infection outcome and should be more closely investigated.
Collapse
Affiliation(s)
- Fernanda K. Barreto
- Laboratório Hematologia, Genética e Biologia Computacional, Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Felipe F.A. Rego
- Laboratório Hematologia, Genética e Biologia Computacional, Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Loianna M. Fonseca
- Laboratório Hematologia, Genética e Biologia Computacional, Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | | | - Thessika H.A. Araújo
- Laboratório Hematologia, Genética e Biologia Computacional, Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Aline Cristina A. Mota-Miranda
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Instituto de Ciências da Saúde, Salvador, Bahia, Brazil
| | - Joana P. Monteiro-Cunha
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Instituto de Ciências da Saúde, Salvador, Bahia, Brazil
| | - Luiz Carlos J. Alcantara
- Laboratório Hematologia, Genética e Biologia Computacional, Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| |
Collapse
|
18
|
Lairmore MD. Animal models of bovine leukemia virus and human T-lymphotrophic virus type-1: insights in transmission and pathogenesis. Annu Rev Anim Biosci 2013; 2:189-208. [PMID: 25384140 DOI: 10.1146/annurev-animal-022513-114117] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bovine leukemia virus (BLV) and human T-lymphotrophic virus type-1 (HTLV-1) are related retroviruses associated with persistent and lifelong infections and a low incidence of lymphomas within their hosts. Both viruses can be spread through contact with bodily fluids containing infected cells, most often from mother to offspring through breast milk. Each of these complex retroviruses contains typical gag, pol, and env genes but also unique, nonstructural proteins encoded from the pX region. These nonstructural genes encode the Tax and Rex regulatory proteins, as well as novel proteins essential for viral spread in vivo. Improvements in the molecular tools to test these viral determinants in cellular and animal models have provided new insights into the pathogenesis of each virus. Comparisons of BLV and HTLV-1 provide insights into mechanisms of spread and tumor formation, as well as potential approaches to therapeutic intervention against the infections.
Collapse
Affiliation(s)
- Michael D Lairmore
- School of Veterinary Medicine, University of California, Davis, California, 95616;
| |
Collapse
|
19
|
Moratorio G, Fischer S, Bianchi S, Tomé L, Rama G, Obal G, Carrión F, Pritsch O, Cristina J. A detailed molecular analysis of complete bovine leukemia virus genomes isolated from B-cell lymphosarcomas. Vet Res 2013; 44:19. [PMID: 23506507 PMCID: PMC3618307 DOI: 10.1186/1297-9716-44-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 01/30/2013] [Indexed: 01/08/2023] Open
Abstract
It is widely accepted that the majority of cancers result from multiple cellular events leading to malignancy after a prolonged period of clinical latency, and that the immune system plays a critical role in the control of cancer progression. Bovine leukemia virus (BLV) is an oncogenic member of the Retroviridae family. Complete genomic sequences of BLV strains isolated from peripheral blood mononuclear cells (PBMC) from cattle have been previously reported. However, a detailed characterization of the complete genome of BLV strains directly isolated from bovine tumors is much needed in order to contribute to the understanding of the mechanisms of leukemogenesis induced by BLV in cattle. In this study, we performed a molecular characterization of BLV complete genomes from bovine B-cell lymphosarcoma isolates. A nucleotide substitution was found in the glucocorticoid response element (GRE) site of the 5' long terminal repeat (5'LTR) of the BLV isolates. All amino acid substitutions in Tax previously found to be related to stimulate high transcriptional activity of 5'LTR were not found in these studies. Amino acid substitutions were found in the nucleocapsid, gp51 and G4 proteins. Premature stop-codons in R3 were observed. Few mutations or amino acid substitutions may be needed to allow BLV provirus to achieve silencing. Substitutions that favor suppression of viral expression in malignant B cells might be a strategy to circumvent effective immune attack.
Collapse
Affiliation(s)
- Gonzalo Moratorio
- Laboratorio de Virología Molecular, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hajj HE, Nasr R, Kfoury Y, Dassouki Z, Nasser R, Kchour G, Hermine O, de Thé H, Bazarbachi A. Animal models on HTLV-1 and related viruses: what did we learn? Front Microbiol 2012; 3:333. [PMID: 23049525 PMCID: PMC3448133 DOI: 10.3389/fmicb.2012.00333] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 08/28/2012] [Indexed: 12/22/2022] Open
Abstract
Retroviruses are associated with a wide variety of diseases, including immunological, neurological disorders, and different forms of cancer. Among retroviruses, Oncovirinae regroup according to their genetic structure and sequence, several related viruses such as human T-cell lymphotropic viruses types 1 and 2 (HTLV-1 and HTLV-2), simian T cell lymphotropic viruses types 1 and 2 (STLV-1 and STLV-2), and bovine leukemia virus (BLV). As in many diseases, animal models provide a useful tool for the studies of pathogenesis, treatment, and prevention. In the current review, an overview on different animal models used in the study of these viruses will be provided. A specific attention will be given to the HTLV-1 virus which is the causative agent of adult T-cell leukemia/lymphoma (ATL) but also of a number of inflammatory diseases regrouping the HTLV-associated myelopathy/tropical spastic paraparesis (HAM/TSP), infective dermatitis and some lung inflammatory diseases. Among these models, rabbits, monkeys but also rats provide an excellent in vivo tool for early HTLV-1 viral infection and transmission as well as the induced host immune response against the virus. But ideally, mice remain the most efficient method of studying human afflictions. Genetically altered mice including both transgenic and knockout mice, offer important models to test the role of specific viral and host genes in the development of HTLV-1-associated leukemia. The development of different strains of immunodeficient mice strains (SCID, NOD, and NOG SCID mice) provide a useful and rapid tool of humanized and xenografted mice models, to test new drugs and targeted therapy against HTLV-1-associated leukemia, to identify leukemia stem cells candidates but also to study the innate immunity mediated by the virus. All together, these animal models have revolutionized the biology of retroviruses, their manipulation of host genes and more importantly the potential ways to either prevent their infection or to treat their associated diseases.
Collapse
Affiliation(s)
- Hiba El Hajj
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Human T-cell leukemia virus type 1 (HTLV-1) bZIP factor requires cellular transcription factor JunD to upregulate HTLV-1 antisense transcription from the 3' long terminal repeat. J Virol 2012; 86:9070-8. [PMID: 22696638 DOI: 10.1128/jvi.00661-12] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Infection with the human T-cell leukemia virus type 1 (HTLV-1) results in a variety of diseases including adult T-cell leukemia (ATL), a fatal malignancy characterized by the uncontrolled proliferation of virally infected CD4(+) T cells. The HTLV-1 basic leucine zipper factor (HBZ) is believed to contribute to development and maintenance of ATL. Unlike the other HTLV-1 genes, the hbz gene is encoded on the complementary strand of the provirus and therefore is not under direct control of the promoter within the 5' long terminal repeat (LTR) of the provirus. This promoter can undergo inactivating genetic or epigenetic changes during the course of ATL that eliminates expression of all viral genes except that of hbz. In contrast, repressive modifications are not known to occur on the hbz promoter located in the 3' LTR, and hbz expression has been consistently detected in all ATL patient samples. Although Sp1 regulates basal transcription from the HBZ promoter, other factors that activate transcription remain undefined. In this study, we used a proviral reporter construct deleted of the 5' LTR to show that HBZ upregulates its own expression through cooperation with JunD. Activation of antisense transcription was apparent in serum-deprived cells in which the level of JunD was elevated, and elimination of JunD expression by gene knockout or shRNA-mediated knockdown abrogated this effect. Activation through HBZ and JunD additionally required Sp1 binding at the hbz promoter. These data favor a model in which JunD is recruited to the promoter through Sp1, where it heterodimerizes with HBZ thereby enhancing its activity. Separately, hbz gene expression led to an increase in JunD abundance, and this effect correlated with emergence of features of transformed cells in immortalized fibroblasts. Overall, our results suggest that JunD represents a novel therapeutic target for the treatment of ATL.
Collapse
|
22
|
Namiki K, Matsunaga H, Yoshioka K, Tanaka K, Murata K, Ishida J, Sakairi A, Kim J, Tokuhara N, Shibakawa N, Shimizu M, Wada Y, Tokunaga Y, Shigetomi M, Hagihara M, Kimura S, Sudo T, Fukamizu A, Kasuya Y. Mechanism for p38α-mediated experimental autoimmune encephalomyelitis. J Biol Chem 2012; 287:24228-38. [PMID: 22637476 DOI: 10.1074/jbc.m111.338541] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One of the mitogen-activated protein kinases, p38, has been found to play a crucial role in various inflammatory responses. In this study, we analyzed the roles of p38α in multiple sclerosis, using an animal model, experimental autoimmune encephalomyelitis (EAE). p38α(+/-) mice (p38α(-/-) showed embryonic lethality) showed less severe neurological signs than WT mice. Adoptive transfer of lymph node cells (LNC) from sensitized WT mice with MOG(35-55) to naive WT-induced EAE was much more severe compared with the case using LNC from sensitized p38α(+/-) mice. Comprehensive analysis of cytokines from MOG(35-55)-challenged LNC by Western blot array revealed that production of IL-17 was significantly reduced by a single copy disruption of the p38α gene or a p38 inhibitor. Likewise, by a luciferase reporter assay, an electrophoresis mobility shift assay, and characterization of the relationship between p38 activity and IL-17 mRNA expression, we confirmed that p38 positively regulates transcription of the Il17 gene. Furthermore, oral administration of a highly specific p38α inhibitor (UR-5269) to WT mice at the onset of EAE markedly suppressed the progression of EAE compared with a vehicle group. These results suggest that p38α participates in the pathogenesis of EAE through IL-17 induction.
Collapse
Affiliation(s)
- Kana Namiki
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Colin L, Dekoninck A, Reichert M, Calao M, Merimi M, Van den Broeke A, Vierendeel V, Cleuter Y, Burny A, Rohr O, Van Lint C. Chromatin disruption in the promoter of bovine leukemia virus during transcriptional activation. Nucleic Acids Res 2011; 39:9559-73. [PMID: 21890901 PMCID: PMC3239207 DOI: 10.1093/nar/gkr671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Bovine leukemia virus expression relies on its chromatin organization after integration into the host cell genome. Proviral latency, which results from transcriptional repression in vivo, represents a viral strategy to escape the host immune system and likely allows for tumor progression. Here, we discriminated two types of latency: an easily reactivable latent state of the YR2 provirus and a ‘locked’ latent state of the L267 provirus. The defective YR2 provirus was characterized by the presence of nuclease hypersensitive sites at the U3/R junction and in the R/U5 region of the 5′-long terminal repeat (5′-LTR), whereas the L267 provirus displayed a closed chromatin configuration at the U3/R junction. Reactivation of viral expression in YR2 cells by the phorbol 12-myristate 13-acetate (PMA) plus ionomycin combination was accompanied by a rapid but transient chromatin remodeling in the 5′-LTR, leading to an increased PU.1 and USF-1/USF-2 recruitment in vivo sustained by PMA/ionomycin-mediated USF phosphorylation. In contrast, viral expression was not reactivated by PMA/ionomycin in L267 cells, because the 5′-LTR U3/R region remained inaccessible to nucleases and hypermethylated at CpG dinucleotides. Remarkably, we elucidated the BLV 5′-LTR chromatin organization in PBMCs isolated from BLV-infected cows, thereby depicting the virus hiding in vivo in its natural host.
Collapse
Affiliation(s)
- Laurence Colin
- Laboratoire de Virologie Moléculaire, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, Rue des Profs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Rodríguez SM, Florins A, Gillet N, de Brogniez A, Sánchez-Alcaraz MT, Boxus M, Boulanger F, Gutiérrez G, Trono K, Alvarez I, Vagnoni L, Willems L. Preventive and therapeutic strategies for bovine leukemia virus: lessons for HTLV. Viruses 2011; 3:1210-48. [PMID: 21994777 PMCID: PMC3185795 DOI: 10.3390/v3071210] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 01/06/2023] Open
Abstract
Bovine leukemia virus (BLV) is a retrovirus closely related to the human T-lymphotropic virus type 1 (HTLV-1). BLV is a major animal health problem worldwide causing important economic losses. A series of attempts were developed to reduce prevalence, chiefly by eradication of infected cattle, segregation of BLV-free animals and vaccination. Although having been instrumental in regions such as the EU, these strategies were unsuccessful elsewhere mainly due to economic costs, management restrictions and lack of an efficient vaccine. This review, which summarizes the different attempts previously developed to decrease seroprevalence of BLV, may be informative for management of HTLV-1 infection. We also propose a new approach based on competitive infection with virus deletants aiming at reducing proviral loads.
Collapse
Affiliation(s)
- Sabrina M. Rodríguez
- Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège (ULg), 4000, Liège, Belgium; E-Mails: (S.M.R.); (N.G.); (F.B.)
| | - Arnaud Florins
- Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), 5030, Gembloux, Belgium; E-Mails: (A.F.); (A.d.B.); (M.T.S.-A.); (M.B.)
| | - Nicolas Gillet
- Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège (ULg), 4000, Liège, Belgium; E-Mails: (S.M.R.); (N.G.); (F.B.)
| | - Alix de Brogniez
- Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), 5030, Gembloux, Belgium; E-Mails: (A.F.); (A.d.B.); (M.T.S.-A.); (M.B.)
| | - María Teresa Sánchez-Alcaraz
- Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), 5030, Gembloux, Belgium; E-Mails: (A.F.); (A.d.B.); (M.T.S.-A.); (M.B.)
| | - Mathieu Boxus
- Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), 5030, Gembloux, Belgium; E-Mails: (A.F.); (A.d.B.); (M.T.S.-A.); (M.B.)
| | - Fanny Boulanger
- Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège (ULg), 4000, Liège, Belgium; E-Mails: (S.M.R.); (N.G.); (F.B.)
| | - Gerónimo Gutiérrez
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA, C.C. 1712, Castelar, Argentina; E-Mails: (G.G.); (K.T.); (I.A.); (L.V.)
| | - Karina Trono
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA, C.C. 1712, Castelar, Argentina; E-Mails: (G.G.); (K.T.); (I.A.); (L.V.)
| | - Irene Alvarez
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA, C.C. 1712, Castelar, Argentina; E-Mails: (G.G.); (K.T.); (I.A.); (L.V.)
| | - Lucas Vagnoni
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA, C.C. 1712, Castelar, Argentina; E-Mails: (G.G.); (K.T.); (I.A.); (L.V.)
| | - Luc Willems
- Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège (ULg), 4000, Liège, Belgium; E-Mails: (S.M.R.); (N.G.); (F.B.)
- Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), 5030, Gembloux, Belgium; E-Mails: (A.F.); (A.d.B.); (M.T.S.-A.); (M.B.)
| |
Collapse
|
25
|
Suntz M, Failing K, Hecht W, Schwartz D, Reinacher M. High prevalence of non-productive FeLV infection in necropsied cats and significant association with pathological findings. Vet Immunol Immunopathol 2010; 136:71-80. [PMID: 20398945 PMCID: PMC7112630 DOI: 10.1016/j.vetimm.2010.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 02/19/2010] [Accepted: 02/19/2010] [Indexed: 12/29/2022]
Abstract
Applying a combination of semi-nested PCR and immunohistology (IHC), the presence of exogenous feline leukemia virus infection was studied in 302 necropsied cats with various disorders. 9% showed the classical outcome of persistent productive FeLV infection which was represented by FeLV antigen expression in different organs. 152 cats (50%) harboured exogenous FeLV-specific proviral sequences in the bone marrow but did not express viral antigen. These cats were considered as horizontally but non-productively infected. Statistical evaluation showed a significant association of non-productive horizontal FeLV infection with a variety of parameters. Non-productively infected cats were statistically significantly older and more often originated from animal shelters than cats without exogenous FeLV infection. Furthermore, some pathological disorders like anemia, panleukopenia, and purulent inflammation showed significant association with non-productive FeLV infection. No significant association was found with lymphosarcoma, known for a long time to be induced by productive FeLV infection.
Collapse
Affiliation(s)
- M Suntz
- Institut für Veterinaer-Pathologie, Justus-Liebig-Universitaet Giessen, Frankfurter Strasse 96, 35392 Giessen, Hessen, Germany.
| | | | | | | | | |
Collapse
|
26
|
Pomier C, Alcaraz MTS, Debacq C, Lançon A, Kerkhofs P, Willems L, Wattel E, Mortreux F. A dose-effect relationship for deltaretrovirus-dependent leukemogenesis in sheep. Retrovirology 2009; 6:30. [PMID: 19344505 PMCID: PMC2670259 DOI: 10.1186/1742-4690-6-30] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 04/03/2009] [Indexed: 01/07/2023] Open
Abstract
Background Retrovirus-induced tumors develop in a broad range of frequencies and after extremely variable periods of time, from only a few days to several decades, depending mainly on virus type. For hitherto unexplained reasons, deltaretroviruses cause hematological malignancies only in a minority of naturally infected organisms and after a very prolonged period of clinical latency. Results Here we demonstrate that the development of malignancies in sheep experimentally infected with the deltaretrovirus bovine leukemia virus (BLV) depends only on the level of BLV replication. Animals were experimentally infected with leukemogenic or attenuated, but infectious, BLV molecular clones and monitored prospectively through 8 months for viral replication. As early as 2 weeks after infection and subsequently at any time during follow-up, leukemogenic viruses produced significantly higher absolute levels of reverse transcription (RT), clonal expansion of infected cells, and circulating proviruses with RT- and somatic-dependent mutations than attenuated viruses. These differences were only quantitative, and both kinds of viruses triggered parallel temporal fluctuations of host lymphoid cells, viral loads, infected cell clonality and proliferation. Conclusion Deltaretrovirus-associated leukemogenesis in sheep appears to be a two-hit process over time depending on the amounts of first horizontally and then vertically expanded viruses.
Collapse
Affiliation(s)
- Carole Pomier
- CNRS FRE-3011-Université Lyon I, Oncovirologie et Biothérapies, Centre Léon Bérard, Lyon, France.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Florins A, Gillet N, Boxus M, Kerkhofs P, Kettmann R, Willems L. Even attenuated bovine leukemia virus proviruses can be pathogenic in sheep. J Virol 2007; 81:10195-200. [PMID: 17626096 PMCID: PMC2045414 DOI: 10.1128/jvi.01058-07] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Based on a reverse genetics approach, we previously reported that bovine leukemia virus (BLV) mutants harboring deletions in the accessory R3 and G4 genes persist at very low proviral loads and are unable to induce leukemia or lymphoma in sheep, indicating that these R3 and G4 gene sequences are required for pathogenesis. We now show that lymphoma can occur, albeit infrequently (1 case of 20) and after extended periods of latency (7 years). Direct sequencing and reinfection experiments demonstrated that lymphomagenesis was not due to the reversion of the mutant to the wild type. Similar observations with another type of attenuated mutant impaired in the transmembrane protein (TM) YXXL signaling motifs were made. We conclude that the R3 and G4 genes and the TM YXXL motifs are not strictly required for pathogenesis but that their integrity contributes to disease frequency and latency.
Collapse
Affiliation(s)
- Arnaud Florins
- Molecular and Cellular Biology Laboratory, National Fund for Scientific Research-FUSAGx, 13 avenue Maréchal Juin, 5030 Gembloux, Belgium
| | | | | | | | | | | |
Collapse
|
28
|
Gillet N, Florins A, Boxus M, Burteau C, Nigro A, Vandermeers F, Balon H, Bouzar AB, Defoiche J, Burny A, Reichert M, Kettmann R, Willems L. Mechanisms of leukemogenesis induced by bovine leukemia virus: prospects for novel anti-retroviral therapies in human. Retrovirology 2007; 4:18. [PMID: 17362524 PMCID: PMC1839114 DOI: 10.1186/1742-4690-4-18] [Citation(s) in RCA: 242] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 03/16/2007] [Indexed: 12/15/2022] Open
Abstract
In 1871, the observation of yellowish nodules in the enlarged spleen of a cow was considered to be the first reported case of bovine leukemia. The etiological agent of this lymphoproliferative disease, bovine leukemia virus (BLV), belongs to the deltaretrovirus genus which also includes the related human T-lymphotropic virus type 1 (HTLV-1). This review summarizes current knowledge of this viral system, which is important as a model for leukemogenesis. Recently, the BLV model has also cast light onto novel prospects for therapies of HTLV induced diseases, for which no satisfactory treatment exists so far.
Collapse
Affiliation(s)
- Nicolas Gillet
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Arnaud Florins
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Mathieu Boxus
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Catherine Burteau
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Annamaria Nigro
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Fabian Vandermeers
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Hervé Balon
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Amel-Baya Bouzar
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Julien Defoiche
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Arsène Burny
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | | | - Richard Kettmann
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | - Luc Willems
- Molecular and Cellular Biology, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
- Luc Willems, National fund for Scientific Research, Molecular and Cellular Biology laboratory, 13 avenue Maréchal Juin, 5030 Gembloux, Belgium
| |
Collapse
|
29
|
Zhao X, Jimenez C, Sentsui H, Buehring GC. Sequence polymorphisms in the long terminal repeat of bovine leukemia virus: evidence for selection pressures in regulatory sequences. Virus Res 2006; 124:113-24. [PMID: 17123656 DOI: 10.1016/j.virusres.2006.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 10/15/2006] [Accepted: 10/20/2006] [Indexed: 10/23/2022]
Abstract
Bovine leukemia virus (BLV) is an oncogenic virus widespread in cattle. It belongs to the genus Deltaretrovirus of the family Retroviridae along with human and simian T-lymphotropic viruses. The BLV transcriptional promoter is located in the proviral 5' long terminal repeat (LTR), composed of U3, R, and U5 regions. BLV LTR contains multiple cis-acting elements important for promoter activity, a short coding sequence (encoding the NH(2) terminus of the G4 regulatory protein), and non-regulatory/non-coding regions. Variation in coding sequences of BLV structural proteins has been studied extensively, but little work has been done on sequence variability of non-coding regions, mostly located in LTR. Here, we report the first study on the natural diversity of the BLV LTR, using viral isolates from 52 cattle in several different areas worldwide. Nucleotide variations from the consensus sequence were observed in most isolates and clustered phylogenetically, corresponding to the geographic distribution of donor cattle. Overall, regulatory regions were significantly more conserved than non-regulatory regions in the BLV LTR, as well as in LTR sub-regions (U3, R, and U5). Evidence of selection pressures in BLV LTR suggests that selection occurs not only in coding sequences, but may also involve regulatory sequences.
Collapse
Affiliation(s)
- Xiangrong Zhao
- Graduate Program in Endocrinology, University of California, Berkeley, CA 94720-3140, USA.
| | | | | | | |
Collapse
|
30
|
Hartl M, Karagiannidis AI, Bister K. Cooperative cell transformation by Myc/Mil(Raf) involves induction of AP-1 and activation of genes implicated in cell motility and metastasis. Oncogene 2006; 25:4043-55. [PMID: 16491116 DOI: 10.1038/sj.onc.1209441] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 01/11/2006] [Accepted: 01/12/2006] [Indexed: 01/26/2023]
Abstract
Avian fibroblasts transformed simultaneously by the v-myc and v-mil(raf) oncogenes of acute leukemia and carcinoma virus MH2 contain elevated levels of c-Fos and c-Jun, major components of the transcription factor complex AP-1. To define specific transcriptional targets in these cells, subtractive hybridization techniques were employed leading to the identification of strongly upregulated genes including OPN (osteopontin), 126MRP, and rac2. OPN is a cytokine and cell attachment protein which has been implicated in human tumor progression and metastasis, the calcium binding 126MRP protein is related to the human S100 protein family involved in invasive cell growth, and the Rac2 protein belongs to the Rho family of small GTPases regulating actin reorganization and cell migration. Promoter analysis indicated that OPN activation is mediated by a non-consensus AP-1 binding site located close to the transcription start site. Electrophoretic mobility shift assays, chromatin immunoprecipitation and transcriptional reporter gene analyses showed that c-Fos and c-Jun bind specifically to this site and that c-Fos efficiently transactivates the OPN promoter. High-level expression of OPN, 126MRP, or Rac2 proteins from a retroviral vector led to partial cell transformation, documented by morphological changes and anchorage-independent growth. The specific activation in v-myc/v-mil(raf)-transformed cells of target genes with intrinsic oncogenic potential may provide an explanation for the longstanding observation that concomitant expression of these oncogenes leads to strongly enhanced oncogenicity in vivo and in vitro compared to cell transformation by v-myc or v-mil(raf) alone.
Collapse
Affiliation(s)
- M Hartl
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| | | | | |
Collapse
|
31
|
van den Heuvel MJ, Jefferson BJ, Jacobs RM. Purified bovine plasma blocking factor decreases Bovine leukemia virus p24 expression while increasing protein synthesis and transcriptional activity of peripheral blood mononuclear cells in short-term culture. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2005; 69:186-92. [PMID: 16187548 PMCID: PMC1176297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Bovine leukemia virus (BLV) induces a persistent infection in the B-cells causing polyclonal expansion of B-cells in one-third of infected cattle and lymphosarcoma in less than 5% of infected cattle. While BLV is difficult to detect in vivo, it is readily produced by cultured lymphocytes and is diminished when supplemented by bovine plasma. This phenomenon is attributed to a poorly characterized plasma blocking factor (PBF). We assessed the effects of bovine plasma on cell viability and BLV p24 expression, and the effects of purified PBF on protein synthesis and gene expression of short-term cultures of bovine lymphocytes. The addition of 25% plasma or semi-purified PBF to cultures had no significant effect on cell viability but caused significant decreases in BLV p24 production and significantly increased de novo protein synthesis. Utilizing a human microarray, the RNA messages of 83 genes involved in cell division, cell metabolism, and gene regulation were up-regulated.
Collapse
|
32
|
Miller MM, Jarosinski KW, Schat KA. Positive and negative regulation of chicken anemia virus transcription. J Virol 2005; 79:2859-68. [PMID: 15709005 PMCID: PMC548473 DOI: 10.1128/jvi.79.5.2859-2868.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Chicken anemia virus (CAV) is a small circular single-stranded DNA virus with a single promoter-enhancer region containing four consensus cyclic AMP response element sequences (AGCTCA), which are similar to the estrogen response element (ERE) consensus half-sites (A)GGTCA. These sequences are arranged as direct repeats, an arrangement that can be recognized by members of the nuclear receptor superfamily. Transient-transfection assays which use a short CAV promoter construct that ended at the transcription start site and drive expression of enhanced green fluorescent protein (EGFP) showed high basal activity in DF-1, LMH, LMH/2A, and primary theca and granulosa cells. The estrogen receptor-enhanced cell line, LMH/2A, had significantly greater expression than LMH cells, and this expression was significantly increased with estrogen treatment. A long promoter construct which included GGTCA-like sequences downstream of the first CAV protein translation start site was found to have significantly less EGFP expression in DF-1 cells than the short promoter, which was largely due to decreased RNA transcription. DNA-protein binding assays indicated that proteins recognizing a consensus ERE palindrome also bind GGTCA-like sequences in the CAV promoter. Estrogen receptor and other members of the nuclear receptor superfamily may provide a mechanism to regulate CAV activity in situations of low virus copy number.
Collapse
Affiliation(s)
- Myrna M Miller
- Unit of Avian Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
33
|
Calomme C, Dekoninck A, Nizet S, Adam E, Nguyên TLA, Van Den Broeke A, Willems L, Kettmann R, Burny A, Van Lint C. Overlapping CRE and E box motifs in the enhancer sequences of the bovine leukemia virus 5' long terminal repeat are critical for basal and acetylation-dependent transcriptional activity of the viral promoter: implications for viral latency. J Virol 2004; 78:13848-64. [PMID: 15564493 PMCID: PMC533944 DOI: 10.1128/jvi.78.24.13848-13864.2004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Accepted: 08/04/2004] [Indexed: 11/20/2022] Open
Abstract
Bovine leukemia virus (BLV) infection is characterized by viral latency in a large proportion of cells containing an integrated provirus. In this study, we postulated that mechanisms directing the recruitment of deacetylases to the BLV 5' long terminal repeat (LTR) could explain the transcriptional repression of viral expression in vivo. Accordingly, we showed that BLV promoter activity was induced by several deacetylase inhibitors (such as trichostatin A [TSA]) in the context of episomal LTR constructs and in the context of an integrated BLV provirus. Moreover, treatment of BLV-infected cells with TSA increased H4 acetylation at the viral promoter, showing a close correlation between the level of histone acetylation and transcriptional activation of the BLV LTR. Among the known cis-regulatory DNA elements located in the 5' LTR, three E box motifs overlapping cyclic AMP responsive elements (CREs) in U3 were shown to be involved in transcriptional repression of BLV basal gene expression. Importantly, the combined mutations of these three E box motifs markedly reduced the inducibility of the BLV promoter by TSA. E boxes are susceptible to recognition by transcriptional repressors such as Max-Mad-mSin3 complexes that repress transcription by recruiting deacetylases. However, our in vitro binding studies failed to reveal the presence of Mad-Max proteins in the BLV LTR E box-specific complexes. Remarkably, TSA increased the occupancy of the CREs by CREB/ATF. Therefore, we postulated that the E box-specific complexes exerted their negative cooperative effect on BLV transcription by steric hindrance with the activators CREB/ATF and/or their transcriptional coactivators possessing acetyltransferase activities. Our results thus suggest that the overlapping CRE and E box elements in the BLV LTR were selected during evolution as a novel strategy for BLV to allow better silencing of viral transcription and to escape from the host immune response.
Collapse
Affiliation(s)
- Claire Calomme
- Université Libre de Bruxelles (ULB), Institut de Biologie et de Médecine Moléculaires (IBMM), Service de Chimie Biologique, Laboratoire de Virologie Moléculaire, Rue des Profs Jeener et Brachet, 12, 6041 Gosselies, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Debacq C, Sanchez Alcaraz MT, Mortreux F, Kerkhofs P, Kettmann R, Willems L. Reduced proviral loads during primo-infection of sheep by Bovine Leukemia virus attenuated mutants. Retrovirology 2004; 1:31. [PMID: 15462678 PMCID: PMC526217 DOI: 10.1186/1742-4690-1-31] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2004] [Accepted: 10/05/2004] [Indexed: 11/23/2022] Open
Abstract
Background The early stages consecutive to infection of sheep (e.g. primo-infection) by Bovine leukemia virus mutants are largely unknown. In order to better understand the mechanisms associated with this period, we aimed at analyzing simultaneously three parameters: B-lymphocytosis, cell proliferation and viral replication. Results Sheep were experimentally infected either with a wild type BLV provirus or with selected mutants among which: a virus harboring an optimalized LTR promoter with consensus cyclic AMP-responsive elements, two deletants of the R3 or the G4 accessory genes and a fusion-deficient transmembrane recombinant. Seroconversion, as revealed by the onset of an anti-viral antibody response, was detected at 3 to 11 weeks after inoculation. At seroconversion, all sheep exhibited a marked increase in the numbers of circulating B lymphocytes expressing the CD5 and CD11b cluster of differentiation markers and, interestingly, this phenomenon occurred independently of the type of virus. The net increase of the absolute number of B cells was at least partially due to accelerated proliferation as revealed, after intravenous injection of bromodeoxyuridine, by the higher proportion of circulating BrdU+ B lymphocytes. BLV proviral DNA was detected by polymerase chain reaction in the leucocytes of all sheep, as expected. However, at seroconversion, the proviral loads were lower in sheep infected by the attenuated proviruses despite similar levels of B cell lymphocytosis. Conclusions We conclude that the proviral loads are not directly linked to the extent of B cell proliferation observed during primo-infection of BLV-infected sheep. We propose a model of opportunistic replication of the virus supported by a general activation process of B lymphocytes.
Collapse
Affiliation(s)
| | | | - Franck Mortreux
- Unité d'Oncogenèse Virale, CNRS UMR5537, Centre Léon Bérard, Lyon, France
| | - Pierre Kerkhofs
- Department of Virology, Veterinary and Agrochemical Research Centre, Uccle, Belgium
| | | | - Luc Willems
- Molecular and cellular biology, FUSAGx, Gembloux, Belgium
| |
Collapse
|
35
|
Harms JS, Eakle KA, Kuo LS, Bremel RD, Splitter GA. Comparison of bovine leukemia virus (BLV) and CMV promoter-driven reporter gene expression in BLV-infected and non-infected cells. GENETIC VACCINES AND THERAPY 2004; 2:11. [PMID: 15327692 PMCID: PMC516020 DOI: 10.1186/1479-0556-2-11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Accepted: 08/24/2004] [Indexed: 11/25/2022]
Abstract
Background Viral promoters are used in mammalian expression vectors because they generally have strong activity in a wide variety of cells of differing tissues and species. Methods The utility of the BLV LTR/promoter (BLVp) for use in mammalian expression vectors was investigated through direct comparison to the CMV promoter (CMVp). Promoter activity was measured using luciferase assays of cell lines from different tissues and species stably transduced with BLVp or CMVp driven luciferase vectors including D17, FLK, BL3.1 and primary bovine B cells. Cells were also modified through the addition of BLV Tax expression vectors and/or BLV infection as well as treatment with trichostatin A (TSA). Results Results indicate the BLV promoter, while having low basal activity compared to the CMV promoter, can be induced to high-levels of activity similar to the CMV promoter in all cells tested. Tax or BLV infection specifically enhanced BLVp activity with no effect on CMVp activity. In contrast, the non-specific activator, TSA, enhanced both BLVp and CMVp activity. Conclusion Based on these data, we conclude the BLV promoter could be very useful for transgene expression in mammalian expression vectors.
Collapse
Affiliation(s)
- Jerome S Harms
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706-1581, USA
| | - Kurt A Eakle
- GALA Biotech, 8137 Forsythia Street, Middleton, WI 53562, USA
| | - Lillian S Kuo
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706-1581, USA
| | - Robert D Bremel
- IoGenetics LLC, 3591 Anderson St., Suite 218, Madison, WI 53704, USA
| | - Gary A Splitter
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706-1581, USA
| |
Collapse
|
36
|
Tajima S, Tsukamoto M, Aida Y. Latency of viral expression in vivo is not related to CpG methylation in the U3 region and part of the R region of the long terminal repeat of bovine leukemia virus. J Virol 2003; 77:4423-30. [PMID: 12634400 PMCID: PMC150652 DOI: 10.1128/jvi.77.7.4423-4430.2003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bovine leukemia virus (BLV) is silent in most cells detectable in vivo, and the repression of its expression allows BLV to evade the host's immune response. In this study, we examined whether CpG methylation of DNA might be involved in the regulation of the expression of BLV in vivo. To investigate the effects of CpG methylation on the activity of the long terminal repeat (LTR) of BLV, we measured the transactivation activity of this region after treatment with the CpG methyltransferase SssI by using a luciferase reporter system. The activity of methylated LTR was significantly lower than that of nonmethylated LTR. Therefore, we examined the extent of CpG methylation of the U3 region and part of the R region of the LTR in BLV-infected cattle and in experimentally BLV-infected sheep at various clinical stages by the bisulfite genomic sequencing method. We detected no or minimal CpG methylation at all stages examined in cattle and sheep, and our results indicate that CpG methylation probably does not participate in the silencing of BLV in vivo.
Collapse
Affiliation(s)
- Shigeru Tajima
- Retrovirus Research Unit, RIKEN, Wako, Saitama 351-0198, Japan
| | | | | |
Collapse
|
37
|
Gatot JS, Callebaut I, Van Lint C, Demonté D, Kerkhofs P, Portetelle D, Burny A, Willems L, Kettmann R. Bovine leukemia virus SU protein interacts with zinc, and mutations within two interacting regions differently affect viral fusion and infectivity in vivo. J Virol 2002; 76:7956-67. [PMID: 12134000 PMCID: PMC155115 DOI: 10.1128/jvi.76.16.7956-7967.2002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2002] [Accepted: 05/10/2002] [Indexed: 11/20/2022] Open
Abstract
Bovine leukemia virus (BLV) and human T-cell lymphotropic virus type 1 (HTLV-1) belong to the genus of deltaretroviruses. Their entry into the host cell is supposed to be mediated by interactions of the extracellular (SU) envelope glycoproteins with cellular receptors. To gain insight into the mechanisms governing this process, we investigated the ability of SU proteins to interact with specific ligands. In particular, by affinity chromatography, we have shown that BLV SU protein specifically interacted with zinc ions. To identify the protein domains involved in binding, 16 peptides distributed along the sequence were tested. Two of them appeared to be able to interact with zinc. To unravel the role of these SU regions in the biology of the virus, mutations were introduced into the env gene of a BLV molecular clone in order to modify residues potentially interacting with zinc. The fusogenic capacity of envelope mutated within the first zinc-binding region (104 to 123) was completely abolished. Furthermore, the integrity of this domain was also required for in vivo infectivity. In contrast, mutations within the second zinc-binding region (218 to 237) did not hamper the fusogenic capacity; indeed, the syncytia were even larger. In sheep, mutations in region 218 to 237 did not alter infectivity or viral spread. Finally, we demonstrated that the envelope of the related HTLV-1 was also able to bind zinc. Interestingly, zinc ions were found to be associated with the receptor-binding domain (RBD) of Friend murine leukemia virus (Fr-MLV) SU glycoprotein, further supporting their relevance in SU structure. Based on the sequence similarities shared with the Fr-MLV RBD, whose three-dimensional structure has been experimentally determined, we located the BLV zinc-binding peptide 104-123 on the opposite side of the potential receptor-binding surface. This observation supports the hypothesis that zinc ions could mediate interactions of the SU RBD either with the C-terminal part of SU, thereby contributing to the SU structural integrity, or with a partner(s) different from the receptor.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Binding Sites/genetics
- Cattle
- Cells, Cultured
- Cricetinae
- Cysteine/chemistry
- DNA, Viral/genetics
- Enzootic Bovine Leukosis/etiology
- Gene Products, env/genetics
- Gene Products, env/physiology
- Human T-lymphotropic virus 1/physiology
- Humans
- Leukemia Virus, Bovine/genetics
- Leukemia Virus, Bovine/pathogenicity
- Leukemia Virus, Bovine/physiology
- Membrane Fusion
- Models, Molecular
- Molecular Sequence Data
- Mutation
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Retroviridae Proteins, Oncogenic/genetics
- Retroviridae Proteins, Oncogenic/physiology
- Transfection
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/physiology
- Virulence
- Zinc/metabolism
Collapse
Affiliation(s)
- Jean-Stéphane Gatot
- Unité de Biologie Cellulaire et Moléculaire, Faculté Universitaire des Sciences Agronomiques, Gembloux, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Debacq C, Asquith B, Kerkhofs P, Portetelle D, Burny A, Kettmann R, Willems L. Increased cell proliferation, but not reduced cell death, induces lymphocytosis in bovine leukemia virus-infected sheep. Proc Natl Acad Sci U S A 2002; 99:10048-53. [PMID: 12119390 PMCID: PMC126622 DOI: 10.1073/pnas.142100999] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lymphocyte homeostasis is the result of a critical balance between cell proliferation and death. Disruption of this subtle equilibrium can lead to the onset of leukemia, an increase in the number of lymphocytes being potentially due to both of these parameters. The relative importance of cell proliferation vs. apoptosis during pathogenesis induced by the primate T cell lymphotropic viruses and bovine leukemia virus (BLV) has been difficult to assess because of conflicting data from a range of in vitro and ex vivo experimental systems. Here, we aim to resolve this issue by measuring the rates of cell proliferation and death in the BLV-ovine system, an animal model of human T lymphotropic virus (HTLV-1). We use a method based on the i.v. injection of 5-bromodeoxyuridine into BLV-infected sheep. We show that B lymphocytes in BLV(+) asymptomatic sheep proliferate significantly faster than in uninfected controls (average proliferation rate: 0.020 per day vs. 0.011 per day). In contrast, the rates of cell death were not significantly different between aleukemic BLV-infected and control sheep (average death rate 0.089 per day vs. 0.094 per day, respectively). We conclude that the increase in the number of B cells during BLV-induced lymphocytosis results from higher proliferation rates but is not due to a significant decrease in apoptosis, in contrast to data from in vitro (ex vivo) experiments. The imbalance created by the net increase in proliferation in the absence of compensating cell death reveals a complex mechanism of feedback regulation controlling homeostasis in the blood compartment.
Collapse
Affiliation(s)
- Christophe Debacq
- Department of Applied Biochemistry and Biology, Faculty of Agronomy, 5030 Gembloux, Belgium
| | | | | | | | | | | | | |
Collapse
|
39
|
Merezak C, Reichert M, Van Lint C, Kerkhofs P, Portetelle D, Willems L, Kettmann R. Inhibition of histone deacetylases induces bovine leukemia virus expression in vitro and in vivo. J Virol 2002; 76:5034-42. [PMID: 11967319 PMCID: PMC136152 DOI: 10.1128/jvi.76.10.5034-5042.2002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Packaging into nucleosomes results in a global transcriptional repression as a consequence of exclusion of sequence-specific factors. This inhibition can be relieved by using inhibitors of histone deacetylases, acetylation being a major characteristic of transcriptionally active chromatin. Paradoxically, the expression of only approximately 2% of the total cellular genes is modulated by histone hyperacetylation. To unravel the potential role of this transcriptional control on BLV expression, we tested the effect of two highly specific inhibitors of deacetylases, trichostatin A (TSA) and trapoxin (TPX). Our results demonstrate that treatment with TSA efficiently enhanced long terminal repeat-directed gene expression of integrated reporter constructs in heterologous D17 stable cell lines. To further examine the biological relevance of these observations made in vitro, we analyzed ex vivo-isolated peripheral blood mononuclear cells (PBMCs) from bovine leukemia virus (BLV)-infected sheep. TSA deacetylase inhibitor induced a drastic increase in viral expression at levels comparable to those induced by treatment with phorbol-12-myristate 13-acetate and ionomycin, the most efficient activators of BLV expression known to date. TSA acted directly on BLV-infected B lymphocytes to increase viral expression and does not seem to require T-cell cooperation. Inhibition of deacetylation after treatment with TSA or TPX also significantly increased viral expression in PBMCs from cattle, the natural host for BLV. Together, our results show that BLV gene expression is, like that of a very small fraction of cellular genes, also regulated by deacetylation.
Collapse
Affiliation(s)
- C Merezak
- Molecular and Cellular Biology, Faculty of Agronomy, Gembloux, Belgium
| | | | | | | | | | | | | |
Collapse
|