1
|
Noncoding RNAs in Retrovirus Replication. RETROVIRUS-CELL INTERACTIONS 2018. [PMCID: PMC7173536 DOI: 10.1016/b978-0-12-811185-7.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although a limited percentage of the genome produces proteins, approximately 90% is transcribed, indicating important roles for noncoding RNA (ncRNA). It is now known that these ncRNAs have a multitude of cellular functions ranging from the regulation of gene expression to roles as structural elements in ribonucleoprotein complexes. ncRNA is also represented at nearly every step of viral life cycles. This chapter will focus on ncRNAs of both host and viral origin and their roles in retroviral life cycles. Cellular ncRNA represents a significant portion of material packaged into retroviral virions and includes transfer RNAs, 7SL RNA, U RNA, and vault RNA. Initially thought to be random packaging events, these host RNAs are now proposed to contribute to viral assembly and infectivity. Within the cell, long ncRNA and endogenous retroviruses have been found to regulate aspects of the retroviral life cycle in diverse ways. Additionally, the HIV-1 transactivating response element RNA is thought to impact viral infection beyond the well-characterized role as a transcription activator. RNA interference, thought to be an early version of the innate immune response to viral infection, can still be observed in plants and invertebrates today. The ability of retroviral infection to manipulate the host RNAi pathway is described here. Finally, RNA-based therapies, including gene editing approaches, are being explored as antiretroviral treatments and are discussed.
Collapse
|
2
|
Wei WZ, Jones RF, Juhasz C, Gibson H, Veenstra J. Evolution of animal models in cancer vaccine development. Vaccine 2015; 33:7401-7407. [PMID: 26241945 DOI: 10.1016/j.vaccine.2015.07.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/02/2015] [Indexed: 12/29/2022]
Abstract
Advances in cancer vaccine development are facilitated by animal models reflecting key features of human cancer and its interface with host immunity. Several series of transplantable preneoplastic and neoplastic mouse mammary lesions have been used to delineate mechanisms of anti-tumor immunity. Mimicking immune tolerance to tumor-associated antigens (TAA) such as HER2/neu, transgenic mice developing spontaneous mammary tumors are strong model systems for pre-clinical vaccine testing. In these models, HER2 DNA vaccines are easily administered, well-tolerated, and induce both humoral and cellular immunity. Although engineered mouse strains have advanced cancer immunotherapy, basic shortcomings remain. For example, multiple mouse strains have to be tested to recapitulate genetic regulation of immune tolerance in humans. Outbred domestic felines more closely parallel humans in the natural development of HER2 positive breast cancer and their varying genetic background. Electrovaccination with heterologous HER2 DNA induces robust adaptive immune responses in cats. Importantly, homologous feline HER2 DNA with a single amino acid substitution elicits unique antibodies to feline mammary tumor cells, unlocking a new vaccine principle. As an alternative approach to targeted vaccination, non-surgical tumor ablation such as cryoablation induces anti-tumor immunity via in situ immunization, particularly when combined with toll-like receptor (TLR) agonist. As strategies for vaccination advance, non-invasive monitoring of host response becomes imperative. As an example, magnetic resonance imaging (MRI) and positron emission tomography (PET) scanning following administration of tryptophan metabolism tracer [11C]-alpha-methyl-tryptophan (AMT) provides non-invasive imaging of both tumor growth and metabolic activities. Because AMT is a substrate of indoleamine-pyrrole 2,3-dioxygenase (IDO), an enzyme that produces the immune regulatory molecule kynurenine, AMT imaging can provide novel insight of host response. In conclusion, new feline models improve the predictive power of cancer immunotherapy and real-time PET imaging enables mechanistic monitoring of host immunity. Strategic utilization of these new tools will expedite cancer vaccine development.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States.
| | - Richard F Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Csaba Juhasz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Heather Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Jesse Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| |
Collapse
|
3
|
Mertz JA, Chadee AB, Byun H, Russell R, Dudley JP. Mapping of the functional boundaries and secondary structure of the mouse mammary tumor virus Rem-responsive element. J Biol Chem 2009; 284:25642-52. [PMID: 19632991 DOI: 10.1074/jbc.m109.012476] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mouse mammary tumor virus (MMTV) is a complex retrovirus that encodes at least three regulatory and accessory proteins, including Rem. Rem is required for nuclear export of unspliced viral RNA and efficient expression of viral proteins. Our previous data indicated that sequences at the envelope-3' long terminal repeat junction are required for proper export of viral RNA. To further map the Rem-responsive element (RmRE), reporter vectors containing various portions of the viral envelope gene and the 3' long terminal repeat were tested in the presence and absence of Rem in transient transfection assays. A 476-bp fragment that spans the envelope-long terminal repeat junction had activity equivalent to the entire 3'-end of the mouse mammary tumor virus genome, but further deletions at the 5'- or 3'-ends reduced Rem responsiveness. RNase structure mapping of the full-length RmRE and a 3'-truncation suggested multiple domains with local base pairing and intervening single-stranded segments. A secondary structure model constrained by these data is reminiscent of the RNA response elements of other complex retroviruses, with numerous local stem-loops and long-range base pairs near the 5'- and 3'-boundaries, and differs substantially from an earlier model generated without experimental constraints. Covariation analysis provides limited support for basic features of our model. Reporter assays in human and mouse cell lines revealed similar boundaries, suggesting that the RmRE does not require cell type-specific proteins to form a functional structure.
Collapse
Affiliation(s)
- Jennifer A Mertz
- Section of Molecular Genetics and Microbiology, The University of Texas, Austin, Texas 78712-0162, USA
| | | | | | | | | |
Collapse
|
4
|
Acha-Orbea H, Held W, Scarpellino L, Shakhov AN. Mls: A Link Between Immunology and Retrovirology. Int Rev Immunol 2009; 8:327-36. [PMID: 1351535 DOI: 10.3109/08830189209053516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The nature of the mysterious minor lymphocyte stimulating (Mls) antigens has recently been clarified. These molecules which were key elements for our current understanding of immune tolerance, have a strong influence on the mouse immune system and are encoded by the open reading frame (orf) of endogenous and exogenous mouse mammary tumor viruses (MMTV's). The knowledge that these antigens are encoded by cancerogenic retroviruses opens an interdisciplinary approach for understanding the mechanisms of immune responses and immune tolerance, retroviral carcinogenesis, and retroviral strategies for infection.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/immunology
- CD4-Positive T-Lymphocytes/immunology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/immunology
- Minor Lymphocyte Stimulatory Antigens/genetics
- Minor Lymphocyte Stimulatory Antigens/immunology
- Molecular Sequence Data
- Open Reading Frames
- Proviruses/genetics
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- H Acha-Orbea
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | | | | | | |
Collapse
|
5
|
Etkind PR, Stewart AF, Wiernik PH. Mouse mammary tumor virus (MMTV)-like DNA sequences in the breast tumors of father, mother, and daughter. Infect Agent Cancer 2008; 3:2. [PMID: 18307792 PMCID: PMC2277433 DOI: 10.1186/1750-9378-3-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 02/28/2008] [Indexed: 02/08/2023] Open
Abstract
Background The diagnosis of late onset breast cancer in a father, mother, and daughter living in the same house for decades suggested the possibility of an environmental agent as a common etiological factor. Both molecular and epidemiological data have indicated a possible role for the mouse mammary tumor virus (MMTV), the etiological agent of breast cancer in mice, in a certain percentage of human breast tumors. The aim of this study was to determine if MMTV might be involved in the breast cancer of this cluster of three family members. Results MMTV-like envelope (env) and long terminal repeat (LTR) sequences containing the MMTV superantigen gene (sag) were detected in the malignant tissues of all three family members. The amplified env gene sequences were 98.0%–99.6% homologous to the MMTV env sequences found in the GR, C3H, and BR6 mouse strains. The amplified LTR sequences containing sag sequences segregated to specific branches of the MMTV phylogenetic tree and did not form a distinct branch of their own. Conclusion The presence of MMTV-like DNA sequences in the malignant tissues of all three family members suggests the possibility of MMTV as an etiological agent. Phylogenetic data suggest that the MMTV-like DNA sequences are mouse and not human derived and that the ultimate reservoir of MMTV is most likely the mouse. Although the route by which these family members came to be infected with MMTV is unknown, the possibility exists that such infection may have resulted from a shared exposure to mice.
Collapse
Affiliation(s)
- Polly R Etkind
- Our Lady of Mercy Medical Center-Comprehensive Cancer Center, New York Medical College, Bronx, New York, USA.
| | | | | |
Collapse
|
6
|
Swanson I, Jude BA, Zhang AR, Pucker A, Smith ZE, Golovkina TV. Sequences within the gag gene of mouse mammary tumor virus needed for mammary gland cell transformation. J Virol 2006; 80:3215-24. [PMID: 16537589 PMCID: PMC1440402 DOI: 10.1128/jvi.80.7.3215-3224.2006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previously, we identified a group of replication-competent exogenous mouse mammary tumor viruses that failed to induce mammary tumors in susceptible mice. Sequence comparison of tumorigenic and tumor-attenuated virus variants has linked the ability of virus to cause high-frequency mammary tumors to the gag gene. To determine the specific sequences within the gag gene that contribute to tumor induction, we constructed five distinct chimeric viruses that have various amino acid coding sequences of gag derived from a tumor-attenuated virus replaced by those of highly tumorigenic virus and tested these viruses for tumorigenic capacities in virus-susceptible C3H/HeN mice. Comparing the tumorigenic potentials of these viruses has allowed us to map the region responsible for tumorigenesis to a 253-amino-acid region within the CA and NC regions of the Gag protein. Unlike C3H/HeN mice, BALB/cJ mice develop tumors when infected with all viral variants, irrespective of the gag gene sequences. Using genetic crosses between BALB/cJ and C3H/HeN mice, we were able to determine that the mechanism that confers susceptibility to Gag-independent mammary tumors in BALB/cJ mice is inherited as a dominant trait and is controlled by a single gene, called mammary tumor susceptibility (mts), that maps to chromosome 14.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Western
- Cell Transformation, Neoplastic
- Chromosome Mapping
- Chromosomes
- Cloning, Molecular
- Conserved Sequence
- Crosses, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Gene Expression Regulation, Viral
- Genes, gag
- Genetic Engineering
- Haplotypes
- Mammary Neoplasms, Experimental/etiology
- Mammary Neoplasms, Experimental/virology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/pathogenicity
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Transgenic
- Models, Genetic
- Molecular Sequence Data
- Mutagenesis
Collapse
|
7
|
Etkind PR, Stewart AFR, Dorai T, Purcell DJ, Wiernik PH. Clonal isolation of different strains of mouse mammary tumor virus-like DNA sequences from both the breast tumors and non-Hodgkin's lymphomas of individual patients diagnosed with both malignancies. Clin Cancer Res 2005; 10:5656-64. [PMID: 15355890 DOI: 10.1158/1078-0432.ccr-03-0364] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE In a previous study, we had detected the presence of mouse mammary tumor virus (MMTV)-like envelope (ENV) gene sequences in both the breast tumors and non-Hodgkin's lymphoma tissue of two of our breast tumor patients who had been diagnosed simultaneously with both malignancies. The aim of this study was to determine if MMTV-like DNA sequences are present in the breast tumors and non-Hodgkin's lymphomas of additional patients suffering from both malignancies and if so to characterize these sequences in detail. EXPERIMENTAL DESIGN DNA was extracted from formalin-fixed, paraffin-embedded tissue sample blocks of breast tumors and non-Hodgkin's lymphomas from patients suffering from both malignancies. A 250-bp region of the MMTV ENV gene and a 630-bp region of the MMTV long terminal repeat (LTR) open reading frame (ORF) that encodes the MMTV superantigen (sag) gene were amplified by PCR from the isolated DNA. Amplified products were analyzed by Southern blotting, cloned, and sequenced. RESULTS MMTV-like ENV and LTR sequences were detected in both the breast tumors and non-Hodgkin's lymphomas of 6 of 12 patients suffering from both malignancies. A novel mutant of the MMTV ENV gene was identified in these patients. Characterization of the MMTV-like LTR highly variable sag sequences revealed total or nearly total identity to three distinct MMTV proviruses from two different branches of the MMTV phylogenetic tree. CONCLUSIONS The presence of MMTV-like ENV and LTR sequences in both the breast tumors and non-Hodgkin's lymphomas of 6 additional patients suggests a possible involvement of these sequences in these two malignancies. MMTV-like LTR sequence homology to different MMTV proviruses revealed the presence of more than one strain of MMTV-like sequences in each individual suggesting the possibility of multiple infections in these patients.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, Viral/genetics
- Base Sequence
- Blotting, Southern
- Breast Neoplasms/complications
- Breast Neoplasms/virology
- DNA, Viral/genetics
- Female
- Genes, env/genetics
- Humans
- Lymphoma, Non-Hodgkin/complications
- Lymphoma, Non-Hodgkin/virology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/isolation & purification
- Middle Aged
- Molecular Sequence Data
- Open Reading Frames
- Phylogeny
- Polymerase Chain Reaction
- Sequence Homology, Nucleic Acid
- Superantigens/genetics
- Terminal Repeat Sequences
Collapse
Affiliation(s)
- Polly R Etkind
- Our Lady of Mercy Medical Center-Comprehensive Cancer Center, New York Medical College, Bronx, New York, USA
| | | | | | | | | |
Collapse
|
8
|
Selmi C, Ross SR, Ansari AA, Invernizzi P, Podda M, Coppel RL, Gershwin ME. Lack of immunological or molecular evidence for a role of mouse mammary tumor retrovirus in primary biliary cirrhosis. Gastroenterology 2004; 127:493-501. [PMID: 15300582 DOI: 10.1053/j.gastro.2004.05.033] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Recent observations, including a pilot clinical trial, have suggested that a human mouse mammary tumor virus (MMTV) causes primary biliary cirrhosis (PBC). We attempted to confirm such data. METHODS We obtained sera from 101 patients (53 with PBC and 48 controls), fixed liver sections from 10 patients (8 PBC and 2 controls), fresh liver specimens (6 PBC and 6 controls), and fresh peripheral blood lymphocytes (PBLs) (10 PBC and 10 controls). We studied sera for reactivities against 3 different strains of MMTV virions, MMTV(C3H), MMTV(FM), and MMTV(LA), including goat polyclonal antibodies against MMTV virions, gp52, and p27 as positive controls. We stained liver specimens using polyclonal antibodies against MMTV and gp52 and further examined tissue samples and PBLs for specific MMTV genome sequences. RESULTS By Western blot analysis, no detectable reactivity in any of the PBC sera against any of the 3 MMTV strains or MMTV gp52 or p27 was observed. However, viral proteins were recognized by our control positive polyclonal antibodies. We note that 13%-60% of PBC sera presented low reactivity against 2 proteins of approximately 57 and 74 kilodaltons. Such reactivity is related to the trace amounts of mitochondrial antigens in the virus preparations derived from murine mammary tumor tissue. No detectable immunohistochemical or molecular evidence for MMTV was found in the liver specimens or PBLs. CONCLUSIONS We were unable to recapitulate the data on this specific retroviral etiology of PBC and suggest that such data could be the result of contamination.
Collapse
Affiliation(s)
- Carlo Selmi
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California at Davis, 95616, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Faedo M, Ford CE, Mehta R, Blazek K, Rawlinson WD. Mouse Mammary Tumor-Like Virus Is Associated with p53 Nuclear Accumulation and Progesterone Receptor Positivity but not Estrogen Positivity in Human Female Breast Cancer. Clin Cancer Res 2004; 10:4417-9. [PMID: 15240531 DOI: 10.1158/1078-0432.ccr-03-0232] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose is to compare the presence of proteins with known associations with breast cancer-progesterone receptor (PgR), estrogen receptor, and p53, with the prevalence of mouse mammary tumor virus (MMTV)-like DNA sequences in human female breast cancers. EXPERIMENTAL DESIGN A cohort of 128 Australian female breast cancers were screened for MMTV-like DNA sequences using PCR. The presence of PgR, estrogen receptor, and nuclear accumulation of p53 protein was assessed in the same samples using immunohistochemical staining. RESULTS Nuclear accumulation of p53 was significantly more prevalent (P = 0.05) in archival human breast cancers containing MMTV-like DNA sequences. The presence of progesterone receptor was significantly higher in MMTV-positive than MMTV-negative breast cancers (P = 0.01). No correlation between estrogen receptor and MMTV-like DNA sequences was found. CONCLUSIONS MMTV causes breast cancer in mice, and hormones up-regulate expression of virus in mice mammary tissue. It is unknown if this is the case in human breast cancers shown to contain DNA of MMTV-like viruses. The positive association between MMTV-like DNA sequences and PgR indicates hormones and MMTV may play a role in human breast cancer. Mutations of the tumor suppressor gene p53 are common in human breast cancer and are associated with higher grades of cancer. The association of MMTV-like DNA sequences with higher grades of cancer, and the positive association between p53 and MMTV-like DNA sequences clearly warrant additional investigation.
Collapse
Affiliation(s)
- Margaret Faedo
- Virology Division, Department of Microbiology, South Eastern Area Laboratory Services, The Prince of Wales Hospital, Randwick, Australia.
| | | | | | | | | |
Collapse
|
10
|
Gorgette O, Existe A, Boubou MI, Bagot S, Guénet JL, Mazier D, Cazenave PA, Pied S. Deletion of T cells bearing the V beta8.1 T-cell receptor following mouse mammary tumor virus 7 integration confers resistance to murine cerebral malaria. Infect Immun 2002; 70:3701-6. [PMID: 12065512 PMCID: PMC128078 DOI: 10.1128/iai.70.7.3701-3706.2002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2001] [Revised: 01/24/2002] [Accepted: 03/25/2002] [Indexed: 11/20/2022] Open
Abstract
Plasmodium berghei ANKA induces a fatal neurological syndrome known as cerebral malaria (CM) in susceptible mice. Host genetic elements are among the key factors determining susceptibility or resistance to CM. Analysis of mice of the same H-2 haplotype revealed that mouse mammary tumor virus 7 (MTV-7) integration into chromosome 1 is one of the key factors associated with resistance to neurological disease during P. berghei ANKA infection. We investigated this phenomenon by infecting a series of recombinant inbred mice (CXD2), derived from BALB/c (susceptible to CM) and DBA/2 (resistant to CM) mice, with P. berghei ANKA. We observed differences in susceptibility to CM induced by this Plasmodium strain. Mice with the MTV-7 sequence in their genome were resistant to CM, whereas those without integration of this gene were susceptible. Thus, an integrated proviral open reading frame or similar genomic sequences may confer protection against neuropathogenesis during malaria, at least in mice.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Malaria, Cerebral/genetics
- Malaria, Cerebral/immunology
- Male
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Mice, Mutant Strains
- Plasmodium berghei/immunology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes/immunology
- Virus Integration/genetics
- Virus Integration/immunology
Collapse
Affiliation(s)
- Olivier Gorgette
- Unité d'Immunophysiopathologie Infectieuse, CNRS URA 1961, Département d'Immunologie, Institut Pasteur, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Schmitz A, Lund AH, Hansen AC, Duch M, Pedersen FS. Target-cell-derived tRNA-like primers for reverse transcription support retroviral infection at low efficiency. Virology 2002; 297:68-77. [PMID: 12083837 DOI: 10.1006/viro.2002.1380] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reverse transcription of a retroviral genome takes place in the cytoplasm of an infected cell by a process primed by a producer-cell-derived tRNA annealed to an 18-nucleotide primer-binding site (PBS). By an assay involving primer complementation of PBS-mutated vectors we analyzed whether tRNA primers derived from the target cell can sustain reverse transcription during murine leukemia virus (MLV) infection. Transduction efficiencies were 4-5 orders of magnitude below those of comparable producer-cell complementations. However, successful usage of a target-cell-derived tRNA primer was proven by cases of correction of single mismatches between Akv-MLV vectors and complementary tRNA primers toward the primer sequence in the integrated vector. Thus, target-cell-derived tRNA-like primers are able to initiate first-strand cDNA synthesis and plus-strand transfer leading to a complete provirus, suggesting that endogenous tRNAs from the infected cell may also have access to the intracellular viral complex at that step of the replication cycle.
Collapse
Affiliation(s)
- Alexander Schmitz
- Department of Molecular and Structural Biology, Aarhus University, C. F. Moellers Allé, Building 130, Denmark
| | | | | | | | | |
Collapse
|
12
|
Zhu Q, Dudley JP. CDP binding to multiple sites in the mouse mammary tumor virus long terminal repeat suppresses basal and glucocorticoid-induced transcription. J Virol 2002; 76:2168-79. [PMID: 11836394 PMCID: PMC135928 DOI: 10.1128/jvi.76.5.2168-2179.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2001] [Accepted: 11/27/2001] [Indexed: 01/19/2023] Open
Abstract
Mouse mammary tumor virus (MMTV) is transcribed at high levels in the lactating mammary gland to ensure transmission of virus from the milk of infected female mice to susceptible offspring. We previously have shown that the transcription factor CCAAT displacement protein (CDP) is expressed in high amounts in virgin mammary gland, yet DNA-binding activity for the MMTV long terminal repeat (LTR) disappears as mammary tissue differentiates during lactation. CDP is a repressor of MMTV expression and, therefore, MMTV expression is suppressed during early mammary gland development. In this study, we have shown using DNase I footprinting and electrophoretic mobility shift assays that there are at least five CDP-binding sites in the MMTV LTR upstream of those previously described in the promoter-proximal negative regulatory element (NRE). Single mutations in two of these upstream sites (+691 or +692 and +735 relative to the first base of the LTR) reduced CDP binding to the cognate sites and elevated reporter gene expression from the full-length MMTV LTR. Combination of a mutation in the promoter-distal NRE with a mutation in the proximal NRE gave approximately additive increases in LTR-reporter gene activity, suggesting that these binding sites act independently. Mutations in several different CDP-binding sites allowed elevation of reporter gene activity from the MMTV promoter in the absence and presence of glucocorticoids, hormones that contribute to high levels of MMTV transcription during lactation by activation of hormone receptor binding to the LTR. In addition, overexpression of CDP in transient-transfection assays suppressed both basal and glucocorticoid-induced LTR-mediated transcription in a dose-dependent manner. These data suggest that multiple CDP-binding sites contribute independently to regulate binding of positive factors, including glucocorticoid receptor, to the MMTV LTR during mammary gland development.
Collapse
Affiliation(s)
- Quan Zhu
- Section of Molecular Genetics and Microbiology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, 100 W. 24th St., Austin, TX 78705, USA
| | | |
Collapse
|
13
|
Popken-Harris P, Pliml L, Harris L. Sequence and genetic analyses of the 3' terminus and integration sites of the RIII/Sa mouse mammary tumor (MMTV) exogenous provirus. Virus Genes 2002; 23:35-43. [PMID: 11556399 DOI: 10.1023/a:1011175112113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The RIII/Sa mouse mammary tumor virus (MMTV) is one of several exogenous MMTV viruses transmitted to suckling mice through the milk. We report herein the nucleotide sequence of the exogenous RIII/Sa provirus from the central Eco RI site through to the end of the U5 region encoded by the 3' LTR. We also provide a detailed sequence analysis often different 3' terminal exogenous MMTV proviral integration sites within mammary tumor DNA obtained by the technique of genome walking. Using a combination of Southern blotting with 3' end probes and PCR utilizing a unique RIII/Sa specific 3' primer, we confirm that the RIII/Sa provirus integrates multiple times in mouse mammary tumors but that little or no integration occurs in various normal tissues. Southern blotting analyses with 3' end probes also indicate that RIII/Sa mice contain two additional endogenous MMTV loci, mtv-6 and mtv- 17, not previously reported. The combined genetic analyses reported herein distinguish between exogenous proviral integrants and endogenous germline MMTV.
Collapse
Affiliation(s)
- P Popken-Harris
- David F. Hickok Memorial Cancer Research Laboratory, Abbott Northwestern Hospital, the Minneapolis, MN 55407, USA
| | | | | |
Collapse
|
14
|
Abstract
The original model of gene therapy, that of efficient delivery, durable transfer, and stable expression of transgenes to correct a gene defect underlying an inherited disease, is limited in light of improved understanding of the processes involved. Techniques that enable regulated expression of transgenes may enhance safety and allow us to regulate the timing and level of expression with a goal of precisely targeting a therapeutic level between the extremes of suboptimal and supraoptimal thresholds. Using regulated systems to control protein expression has practical and possibly essential roles for the success of safe and effective gene therapy in a number of clinical situations. Pharmacologically regulated gene expression is an evolving tool, and no individual system may be effective in all clinical applications.
Collapse
Affiliation(s)
- P W Zoltick
- Institute for Human Gene Therapy, University of Pennsylvania, Philadelphia 19104, USA
| | | |
Collapse
|
15
|
Modin C, Lund AH, Schmitz A, Duch M, Pedersen FS. Alleviation of murine leukemia virus repression in embryonic carcinoma cells by genetically engineered primer binding sites and artificial tRNA primers. Virology 2000; 278:368-79. [PMID: 11118360 DOI: 10.1006/viro.2000.0683] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The primer binding site (PBS) plays pivotal roles during reverse transcription of retroviruses and also is the target of a cellular host defense impeding the transcription of murine leukemia virus (MLV) harboring a proline (pro) PBS in embryonic cells. Both the PBS and the tRNA primer are copied during reverse transcription and anneal as complementary DNA sequences creating the PBS of the integrated provirus. The pro PBS of MLV can be exchanged by PBS sequences matching endogenous or engineered tRNAs to allow replication of Akv MLV-derived vectors in fibroblasts. Here we use the PBS escape mutant B2 to demonstrate the capacity of the synthetic tRNA(B2) to function in reverse transcription in competition with endogenous tRNAs in fibroblasts and embryonic carcinoma (EC) cells. We further show symmetry between PBS and the primer by the ability of the synthetic tRNA(B2) to confer escape from EC repression of a PBS-Pro vector. Of a panel of vectors with the repressed pro PBS substituted for other natural or artificial PBS sequences, all except one efficiently expressed the neo marker gene when transferred to NIH/3T3 and EC cells, hence avoiding PBS-mediated silencing in EC cells. A non-natural PBS matching an artificially designed tRNA molecule conferred no further relief from repression than that attained with the B2 escape mutant or the natural alternative PBSs. Interestingly, a vector harboring a PBS matching tRNA(Lys1.2) suffered repression similar to the wild-type PBS-Pro but was partially rescued by a single point mutation of the PBS.
Collapse
Affiliation(s)
- C Modin
- Department of Molecular and Structural Biology, University of Aarhus, Aarhus C, DK-8000, Denmark
| | | | | | | | | |
Collapse
|
16
|
Yanagawa S, Lee JS, Kakimi K, Matsuda Y, Honjo T, Ishimoto A. Identification of Notch1 as a frequent target for provirus insertional mutagenesis in T-cell lymphomas induced by leukemogenic mutants of mouse mammary tumor virus. J Virol 2000; 74:9786-91. [PMID: 11000255 PMCID: PMC112415 DOI: 10.1128/jvi.74.20.9786-9791.2000] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In contrast to wild-type mouse mammary tumor virus (MMTV), the MMTV mutants with specific deletions in the U3 region of their long terminal repeats cause T-cell lymphomas. In 30% of T-cell lymphomas arising in BALB/c mice infected with MLA-MMTV, a leukemogenic MMTV mutant, we have found that MMTV proviruses were integrated into a short region of the Notch1 genome, so that truncated Notch1 transcripts encoding the transmembrane and the cytoplasmic domains of Notch1 protein could be expressed. Thus, Notch1 is a major target of provirus insertional mutagenesis in these T-cell lymphomas.
Collapse
Affiliation(s)
- S Yanagawa
- Department of Viral Oncology, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | |
Collapse
|
17
|
Tovar Sepúlveda VA, Berdel B, Coffin JM, Reuss FU. Mouse mammary tumor virus superantigen expression is reduced by glucocorticoid treatment. Virology 2000; 275:98-106. [PMID: 11017791 DOI: 10.1006/viro.2000.0491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Expression of mouse mammary tumor virus (MMTV)-encoded superantigens in B lymphocytes are required for viral transmission and pathogenesis. Due to problems with detection and quantification of the superantigen protein, most reports about the mechanism of superantigen expression from the viral sag gene rely on the quantitative analysis of putative sag mRNAs. The description of multiple promoters as a source of putative sag mRNA has complicated the situation even further. All conclusions about the level of superantigen protein expression based on these data remain circumstantial. To test the effect of the glucocorticoid hormone dexamethasone on the total superantigen expression from an infectious MMTV provirus we used a quantitative assay that is based on a superantigen-luciferase fusion protein. MMTV gene expression from the major promoter in the 5' long terminal repeat (LTR) is strongly induced in the presence of glucocorticoid hormones. We now demonstrate that, in the presence of dexamethasone, sag gene expression is reduced despite increased transcription from the MMTV 5' LTR and increased amounts of putative sag mRNA initiated at the LTR promoter. These data show that the expression of the MMTV sag gene does not correlate with the activity of the major LTR promoter and thus differs from all other MMTV genes.
Collapse
MESH Headings
- Animals
- Cell Line
- Dexamethasone/pharmacology
- Dexamethasone/therapeutic use
- Gene Expression Regulation, Viral/drug effects
- Genes, Reporter/genetics
- Genes, Viral/genetics
- Glucocorticoids/pharmacology
- Glucocorticoids/therapeutic use
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/virology
- Mammary Tumor Virus, Mouse/drug effects
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/immunology
- Mice
- Promoter Regions, Genetic/genetics
- Proviruses/drug effects
- Proviruses/genetics
- Proviruses/immunology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- RNA, Viral/analysis
- RNA, Viral/genetics
- Recombinant Fusion Proteins/genetics
- Superantigens/genetics
- Terminal Repeat Sequences/genetics
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
- Transfection
Collapse
Affiliation(s)
- V A Tovar Sepúlveda
- Deutsches Krebsforschungszentrum, Forschungsschwerpunkt Angewandte Tumorvirologie F0400, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
18
|
Reuss FU, Coffin JM. The mouse mammary tumor virus transcription enhancers for hematopoietic progenitor and mammary gland cells share functional elements. J Virol 2000; 74:8183-7. [PMID: 10933730 PMCID: PMC112353 DOI: 10.1128/jvi.74.17.8183-8187.2000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Expression of mouse mammary tumor virus (MMTV)-encoded superantigens in B lymphocytes is required for viral transmission and pathogenesis. We have previously established a critical role of an enhancer element within the long terminal repeat (LTR) for MMTV sag gene expression in B-lymphoid progenitor cells. We now demonstrate enhancer activity of this element in a promyelocytic progenitor cell line. We also map the position of the enhancer within the U3 region of the MMTV LTR and show that the progenitor cell enhancer shares functional elements with a previously described mammary gland-specific enhancer.
Collapse
Affiliation(s)
- F U Reuss
- Forschungsschwerpunkt Angewandte Tumorvirologie F0400, 69120 Heidelberg, Germany.
| | | |
Collapse
|
19
|
Xu L, Tay CH, Huber BT, Sarkar NH. Cloning of an infectious milk-borne mouse mammary tumor virus (MMTV) DNA from a mammary tumor that developed in an endogenous MMTV-free wild mouse. Virology 2000; 273:325-32. [PMID: 10915603 DOI: 10.1006/viro.2000.0437] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Molecular characterization of infectious mouse mammary tumor viruses (MMTVs) has been hampered due to the problem of cloning a full-length exogenous virus into a plasmid. The present report describes our strategy for obtaining a full-length clone of an exogenous MMTV from a mouse mammary tumor that arose spontaneously in a wild Chinese mouse free of endogenous MMTV and shows that the cloned virus (JYG-MMTV) is expressed in rat RBA cells. Four-week-old C58/J x CBA/CaJ female mice, free of both endogenous and exogenous MMTVs, were injected with virus-secreting RBA cells. The progeny of these mice were bred, and their offspring were tested for the presence of MMTV. These third-generation mice were found to actively produce MMTV that was shed in their milk and transmitted to their offspring. The virus was detected not only in the mammary glands of these young mice, but also in their spleens and bone marrow. These results suggest that our plasmid-cloned exogenous JYG-MMTV is infectious. This virus can now be used effectively in manipulating the various genes of JYG-MMTV and other MMTV strains to understand their structure/function relationships.
Collapse
Affiliation(s)
- L Xu
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | |
Collapse
|
20
|
Zhu Q, Gregg K, Lozano M, Liu J, Dudley JP. CDP is a repressor of mouse mammary tumor virus expression in the mammary gland. J Virol 2000; 74:6348-57. [PMID: 10864645 PMCID: PMC112141 DOI: 10.1128/jvi.74.14.6348-6357.2000] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2000] [Accepted: 04/19/2000] [Indexed: 01/19/2023] Open
Abstract
Mouse mammary tumor virus (MMTV) transcription is highest in the lactating mammary gland but is detectable in a variety of other tissues. Previous results have shown that MMTV expression is suppressed in lymphoid and other tissues through the binding of the homeodomain-containing repressor special AT-rich binding protein 1 to a negative regulatory element (NRE) in the MMTV long terminal repeat (LTR). Another homeoprotein repressor, CCAAT displacement protein (CDP), also binds to the MMTV NRE, but a role for CDP in MMTV transcriptional suppression has not yet been demonstrated. In this paper, we show that the level of CDP decreases during development of the mammary gland and that this decline in CDP level correlates with the known increase in MMTV expression observed during mammary gland differentiation. Moreover, CDP overexpression was able to suppress MMTV LTR-reporter gene activity up to 20-fold in transient-transfection assays of mouse mammary cells. To determine if this effect was due to direct binding of CDP to the promoter-proximal NRE, we performed DNase I protection assays to map two CDP-binding sites from +835 to +845 and +920 to +931 relative to the first base of the LTR. Mutations engineered into each of these sites decreased CDP binding to the proximal NRE, whereas a combination of these mutations further reduced binding. Subsequently, each of these mutations was introduced into the full-length MMTV LTR upstream of the luciferase reporter gene. Analysis of stable transfectants of LTR constructs showed that CDP binding site mutations in the proximal NRE elevated reporter gene expression two- to sixfold compared to wild-type LTR constructs. Thus, MMTV expression increases during mammary gland development, in part due to decreased CDP levels and CDP binding to the LTR. Together, these experiments provide the first evidence that CDP acts as a repressor of MMTV transcription in the mammary gland.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- DNA Footprinting
- DNA, Viral/genetics
- DNA, Viral/physiology
- Deoxyribonuclease I
- Female
- Gene Expression Regulation
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/virology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/metabolism
- Mice
- Mice, Inbred BALB C
- Mutagenesis
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Protein Binding
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Terminal Repeat Sequences
Collapse
Affiliation(s)
- Q Zhu
- Section of Molecular Genetics and Microbiology and Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78705, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
We previously reported that multiparous WAP-TGFalpha transgenic mice develop mammary gland carcinomas with complete incidence. TGFalpha-induced tumors appear stochastically and with relatively long latency, indicating an additional requirement for other genetic alterations. To identify genes that cooperate with TGFalpha in mammary tumorigenesis, we used a retroviral insertion approach featuring a cloned and infectious hybrid MMTV (C3H/Mtv-1; (Shackleford and Varmus, 1988)). Tumor latency was decreased approximately 30% in MMTV-infected WAP-TGFalpha transgenic animals compared to noninfected transgenic controls, and > 30% of the corresponding tumors displayed evidence of integrated C3H/Mtv-1 DNA. PCR-based analyses of DNAs from two virus-infected, transgenic tumors revealed integration of hybrid MMTV in 3' untranslated exons of the Wnt-1 or Wnt-3 oncogenes. Moreover, Northern blots confirmed dramatic induction of Wnt-1 or Wnt-3 transcripts in the respective tumors, indicating that MMTV integration resulted in activated expression of these genes. Semiquantitative RT-PCR analyses showed that overexpression of Wnt-1 or Wnt-3 was a common occurrence in MMTV-infected WAP-TGFalpha tumors, and some noninfected WAP-TGFalpha tumors also showed evidence of elevated Wnt-3 transcripts. Collectively, these results reveal cooperative induction of mammary gland tumorigenesis by simultaneous deregulation of EGF-like (TGFalpha) and Wnt growth factors.
Collapse
Affiliation(s)
- J A Schroeder
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill 27599-7260, USA
| | | | | |
Collapse
|
22
|
Lund AH, Schmitz A, Pedersen FS, Duch M. Identification of a novel human tRNA(Ser(CGA)) functional in murine leukemia virus replication. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1492:264-8. [PMID: 11004500 DOI: 10.1016/s0167-4781(00)00095-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have identified a human tRNA(Ser) isoacceptor matching the UCG codon. The tRNA was discovered via its ability to act in reverse transcription of a murine leukemia virus vector containing a complementary tRNA primer binding site (Lund et al., Nucleic Acids Res., 28 (2000) 791-799). The tRNA(Ser(CGA)) was detected in cell lines of human, monkey and mouse origin. The UCG codon is the most rarely used codon in human genes. The cloned human tRNA(Ser(CGA)) gene encodes an 85 nucleotide, intron-less tRNA, contains a consensus split intragenic promoter and is located at region p21.3-22.2 on chromosome 6. The integrity and functionality of the cloned tRNA(Ser(CGA)) gene was verified by in vitro transcription analysis in HeLa nuclear extracts.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cells, Cultured
- Chromosome Mapping
- Chromosomes, Human, Pair 6
- DNA/analysis
- Genome, Human
- Haplorhini
- HeLa Cells
- Humans
- Leukemia Virus, Murine/physiology
- Mice
- Molecular Sequence Data
- Nucleic Acid Conformation
- RNA, Transfer, Ser/chemistry
- RNA, Transfer, Ser/genetics
- RNA, Transfer, Ser/physiology
- Virus Replication
Collapse
Affiliation(s)
- A H Lund
- Department of Molecular and Structural Biology, University of Aarhus, Denmark
| | | | | | | |
Collapse
|
23
|
Lund AH, Duch M, Pedersen FS. Selection of functional tRNA primers and primer binding site sequences from a retroviral combinatorial library: identification of new functional tRNA primers in murine leukemia virus replication. Nucleic Acids Res 2000; 28:791-9. [PMID: 10637332 PMCID: PMC102544 DOI: 10.1093/nar/28.3.791] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Retroviral reverse transcription is initiated from a cellular tRNA molecule and all known exogenous isolates of murine leukemia virus utilise a tRNA(Pro)molecule. While several studies suggest flexibility in murine leukemia virus primer utilisation, studies on human immunodeficiency virus and avian retro-viruses have revealed evidence of molecular adapt-ation towards the specific tRNA isoacceptor used as replication primer. In this study, murine leukemia virus tRNA utilisation is investigated by in vivo screening of a retroviral vector combinatorial library with randomised primer binding sites. While most of the selected primer binding sites are complementary to the 3'-end of tRNA((Pro)), we also retrieved PBS sequences matching four other tRNA molecules and demonstrate that Akv murine leukemia virus vectors may efficiently replicate using tRNA(Arg(CCU)), tRNA(Phe(GAA))and a hitherto unknown human tRNA(Ser(CGA)).
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Base Sequence
- Bias
- Binding Sites
- Blotting, Northern
- Cell Line
- Gene Library
- Genetic Vectors/genetics
- Humans
- Leukemia Virus, Murine/genetics
- Leukemia Virus, Murine/physiology
- Mice
- Nucleic Acid Hybridization/genetics
- Polymerase Chain Reaction
- RNA/genetics
- RNA, Transfer/genetics
- RNA, Transfer, Arg/genetics
- RNA, Transfer, Phe/genetics
- RNA, Transfer, Ser/genetics
- Random Allocation
- Regulatory Sequences, Nucleic Acid/genetics
- Substrate Specificity
- Virus Replication/genetics
Collapse
Affiliation(s)
- A H Lund
- Department of Molecular and Structural Biology, University of Aarhus, Denmark
| | | | | |
Collapse
|
24
|
Dzuris JL, Zhu W, Kapkov D, Golovkina TV, Ross SR. Expression of mouse mammary tumor virus envelope protein does not prevent superinfection in vivo or in vitro. Virology 1999; 263:418-26. [PMID: 10544114 DOI: 10.1006/viro.1999.9967] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inbred mice expressing endogenous mouse mammary tumor virus envelope proteins can be infected with exogenous virus, and the mammary tumors that develop in these mice usually have many proviruses integrated in their genomes, indicating that this virus is not subject to receptor interference. We show here that transgenic mice expressing an exogenous mouse mammary tumor virus (C3H) envelope protein can still be infected with this virus. Moreover, cultured mammary gland cells expressing the mouse mammary tumor virus (C3H) envelope protein can be superinfected with pseudotyped viruses bearing that same protein. Thus cellular expression of the mouse mammary tumor virus envelope protein does not block superinfection in vivo or in vitro.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Line
- DNA, Viral/analysis
- DNA, Viral/genetics
- Female
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/virology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Transgenic
- Proviruses/genetics
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Retroviridae Infections/metabolism
- Retroviridae Infections/virology
- Superinfection/metabolism
- Superinfection/virology
- Transfection
- Transgenes/genetics
- Transgenes/physiology
- Tumor Virus Infections/metabolism
- Tumor Virus Infections/virology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
- J L Dzuris
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6148, USA
| | | | | | | | | |
Collapse
|
25
|
Lund AH, Schmidt J, Luz A, Sørensen AB, Duch M, Pedersen FS. Replication and pathogenicity of primer binding site mutants of SL3-3 murine leukemia viruses. J Virol 1999; 73:6117-22. [PMID: 10364369 PMCID: PMC112678 DOI: 10.1128/jvi.73.7.6117-6122.1999] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/1998] [Accepted: 04/05/1999] [Indexed: 12/29/2022] Open
Abstract
Retroviral reverse transcription is primed by a cellular tRNA molecule annealed to an 18-bp primer binding site sequence. The sequence of the primer binding site coincides with that of a negatively acting cis element that mediates transcriptional silencing of murine leukemia virus (MLV) in undifferentiated embryonic cells. In this study we test whether SL3-3 MLV can replicate stably using tRNA primers other than the cognate tRNAPro and analyze the effect of altering the primer binding site sequence to match the 3' end of tRNA1Gln, tRNA3Lys, or tRNA1,2Arg in a mouse pathogenicity model. Contrary to findings from cell culture studies of primer binding site-modified human immunodeficiency virus type 1 and avian retroviruses, our findings were that SL3-3 MLV may stably and efficiently replicate with tRNA primers other than tRNAPro. Although lymphoma induction of the SL3-3 Lys3 mutant was significantly delayed relative to that of the wild-type virus, molecular tumor analysis indicated that all the primer binding site-modified viruses induce T-cell lymphomas similar to those induced by the wild-type virus in terms of frequencies of genomic rearrangements within the T-cell receptor beta-chain, the immunoglobulin kappa light chain, and the c-myc locus. Whereas none of the mutants were found to revert to tRNAPro primer utilization, in two tumors resulting from the injection of the SL3-3 Lys3 mutant the primer binding site was altered to match that of a new primer species, tRNA1,2Lys. In addition, recombination with endogenous viruses resulting in the generation of recombinant viruses carrying a glutamine primer binding site was detected in the majority of the tumors induced by the SL3-3 Lys3 mutant as well as in two tumors induced by wild-type SL3-3 and the SL3-3 Arg1,2 mutant.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Binding Sites
- DNA, Viral/analysis
- Leukemia Virus, Murine/genetics
- Leukemia Virus, Murine/pathogenicity
- Leukemia Virus, Murine/physiology
- Leukemia, Experimental/virology
- Mice
- Mutation
- RNA
- RNA, Transfer
- RNA, Transfer, Arg
- RNA, Transfer, Gln
- RNA, Transfer, Lys
- RNA, Viral
- Retroviridae Infections/virology
- Tumor Virus Infections/virology
- Virus Replication
Collapse
Affiliation(s)
- A H Lund
- Department of Molecular and Structural Biology, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
26
|
Acha-Orbea H, Finke D, Attinger A, Schmid S, Wehrli N, Vacheron S, Xenarios I, Scarpellino L, Toellner KM, MacLennan IC, Luther SA. Interplays between mouse mammary tumor virus and the cellular and humoral immune response. Immunol Rev 1999; 168:287-303. [PMID: 10399081 DOI: 10.1111/j.1600-065x.1999.tb01299.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mouse mammary tumor virus has developed strategies to exploit the immune response. It requires vigorous immune stimulation to achieve efficient infection. The infected antigen-presenting cells present a viral superantigen on the cell surface which stimulates strong CD4-mediated T-cell help but CD8 T-cell responses are undetectable. Despite the high frequency of superantigen-reactive T cells, the superantigen-induced immune response is comparable to classical antigen responses in terms of T-cell priming, T-cell-B-cell collaboration as well as follicular and extra-follicular B-cell differentiation. Induction of systemic anergy is observed, similar to classical antigen responses where antigen is administered systemically but does not influence the role of the superantigen-reactive T cells in the maintenance of the chronic germinal center reaction. So far we have been unable to detect a cytotoxic T-cell response to mouse mammary tumor virus peptide antigens or to the superantigen. This might yet represent another step in the viral infection strategy.
Collapse
Affiliation(s)
- H Acha-Orbea
- Ludwing Institute of Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Buggiano V, Goldman A, Nepomnaschy I, Bekinschtein P, Berguer P, Lombardi G, Deroche A, Francisco MV, Piazzon I. Characterization of two infectious mouse mammary tumour viruses: superantigenicity and tumorigenicity. Scand J Immunol 1999; 49:269-77. [PMID: 10102644 DOI: 10.1046/j.1365-3083.1999.00502.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mouse mammary tumour virus (MMTV) is a type B retrovirus that causes mammary tumours in susceptible mice. MMTV encodes a superantigen (SAg) that has the property of stimulating T-cell populations expressing a particular variable region of the T-cell receptor (TCR) beta chain (Vbeta) and needs to be presented in the context of major histocompatibility complex (MHC) class II molecules. Previously, we described two exogenous MMTV, MMTV BALB14, which encodes a superantigen that induces the deletion of Vbeta14+ Tcells, and MMTV BALB2, which encodes a SAg that induces the deletion of Vbeta2+ Tcells. We now describe their biological activity: the deletions involve both CD4+ and CD8+ populations, are progressive and can be detected in blood, lymph nodes and spleen. Such deletions reflect, at least in part, those occurring during intrathymic development. Both BALB2 and BALB14 viral variants are capable of inducing a strong increase of Vbeta-specific T cells in BALB/c mice (I-A+, I-E+). However, when injected into the footpad, their initial stimulatory capacity differs in that the presence of MHC I-E molecules is essential only for the stimulation of Vbeta2+ T cells. Both viral variants are able to induce deletion even in the absence of the I-E molecule in which case, however, deletion appears later and is less pronounced. Both exogenous MMTVs induce, at the end of a year, 30-35% of pregnancy-dependent mammary adenocarcinomas.
Collapse
MESH Headings
- Adenocarcinoma/etiology
- Adenocarcinoma/immunology
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Female
- Genetic Variation
- Mammary Neoplasms, Experimental/etiology
- Mammary Neoplasms, Experimental/immunology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/immunology
- Mammary Tumor Virus, Mouse/pathogenicity
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Pregnancy
- Pregnancy Complications, Neoplastic/etiology
- Pregnancy Complications, Neoplastic/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Retroviridae Infections/etiology
- Retroviridae Infections/immunology
- Superantigens/genetics
- T-Lymphocyte Subsets/immunology
- Tumor Virus Infections/etiology
- Tumor Virus Infections/immunology
Collapse
Affiliation(s)
- V Buggiano
- Division Medicina Experimental, Instituto de Investigaciones Hematologicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Robson ND, Telesnitsky A. Effects of 3' untranslated region mutations on plus-strand priming during moloney murine leukemia virus replication. J Virol 1999; 73:948-57. [PMID: 9882295 PMCID: PMC103914 DOI: 10.1128/jvi.73.2.948-957.1999] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/1998] [Accepted: 10/21/1998] [Indexed: 12/17/2022] Open
Abstract
A conserved purine-rich motif located near the 3' end of retroviral genomes is involved in the initiation of plus-strand DNA synthesis. We mutated sequences both within and flanking the Moloney murine leukemia virus polypurine tract (PPT) and determined the effects of these alterations on viral DNA synthesis and replication. Our results demonstrated that both changes in highly conserved PPT positions and a mutation that left only the cleavage-proximal half of the PPT intact led to delayed replication and reduced the colony-forming titer of replication defective retroviral vectors. A mutation that altered the cleavage proximal half of the PPT and certain 3' untranslated region mutations upstream of the PPT were incompatible with or severely impaired viral replication. To distinguish defects in plus-strand priming from other replication defects and to assess the relative use of mutant and wild-type PPTs, we examined plus-strand priming from an ectopic, secondary PPT inserted in U3. The results demonstrated that the analyzed mutations within the PPT primarily affected plus-strand priming whereas mutations upstream of the PPT appeared to affect both plus-strand priming and other stages of viral replication.
Collapse
Affiliation(s)
- N D Robson
- Department of Microbiology and Immunology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109-0620, USA
| | | |
Collapse
|
29
|
Qin W, Golovkina TV, Peng T, Nepomnaschy I, Buggiano V, Piazzon I, Ross SR. Mammary gland expression of mouse mammary tumor virus is regulated by a novel element in the long terminal repeat. J Virol 1999; 73:368-76. [PMID: 9847341 PMCID: PMC103842 DOI: 10.1128/jvi.73.1.368-376.1999] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Mouse mammary tumor virus (MMTV) infects both lymphoid tissue and lactating mammary gland during its infectious cycle, but some endogenous MMTVs are transcribed only in lymphoid cells. We found a lymphoid cell-specific endogenous MMTV that was converted to a milk-borne, infectious virus through recombination with an exogenously transmitted MMTV. The changed expression pattern correlated with the alteration of a single base pair in the long terminal repeat of the lymphoid cell-specific virus. Transgenic mice with the element from either the milk-borne or lymphoid cell-specific virus upstream of the chloramphenicol acetyltransferase reporter gene showed the same pattern of expression as the virus from which the regulatory sequences were derived. Electrophoretic mobility shift assays with mammary cell extracts showed that the site from the milk-borne virus was preferentially bound by a prolactin-inducible factor that poorly bound the altered site from the lymphoid cell-specific virus. The complex that formed on the milk-borne virus-specific oligonucleotide supershifted with anti-Stat5b antibody. Mice lacking either Stat5a or Stat5b had dramatically reduced levels of MMTV transcripts in mammary gland but not in lymphoid tissue. Thus, a member of the STAT family of transcription factors is involved in the tissue-specific expression of mouse mammary tumor virus in vivo. This is the first example of the involvement of a member of the STAT family of transcription factors in the control of tissue-specific expression.
Collapse
Affiliation(s)
- W Qin
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6142, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Golovkina TV, Dudley JP, Ross SR. B and T Cells Are Required for Mouse Mammary Tumor Virus Spread Within the Mammary Gland. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.5.2375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Mouse mammary tumor virus (MMTV) is an infectious retrovirus transmitted through milk from mother to newborns. MMTV encodes a superantigen (SAg) whose activity is indispensable for the virus life cycle, since a genetically engineered virus with a mutation in the sag gene neither amplified in cells of the immune system of suckling pups nor infected their mammary glands. When wild-type MMTV was injected directly into the mammary glands of uninfected pubescent mice, their lymphoid as well as mammary gland cells became virus infected. To test whether this infection of lymphoid cells was dependent on SAg activity and required for virus spread within the mammary gland, we performed mammary gland injections of wild-type MMTV(C3H) into two strains of transgenic mice that lacked SAg-cognate, Vβ14+ T cells. Neither the MTV-ORF or LEL strains showed infection of their mammary glands. Moreover, no MMTV infection of their peripheral lymphocytes was detected. Similar experiments with mice lacking B cells (μ-chain knockouts) showed no detectable virus spread in the mammary glands or lymphoid tissues. These data suggest that SAg activity and MMTV-infected lymphocytes are required, not only for initial steps of viral infection, but also for virus spread within the mammary gland. Virus spread at late times in infection determines whether MMTV induces mammary tumors.
Collapse
Affiliation(s)
- Tatyana V. Golovkina
- *Department of Microbiology/Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| | | | - Susan R. Ross
- *Department of Microbiology/Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| |
Collapse
|
31
|
Schirrmacher V, Beutner U, Bucur M, Umansky V, Rocha M, von Hoegen P. Loss of Endogenous Mouse Mammary Tumor Virus Superantigen Increases Tumor Resistance. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.2.563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
From a cross between a tumor-susceptible mouse strain (DBA/2; D) and a tumor-resistant MHC-identical strain (B10.D2; D2) new recombinant inbred mouse strains were established over many generations of inbreeding and tumor resistance selection. Since resistance to the highly metastatic DBA/2 lymphoma variant ESb had an immunologic basis, and the two parental strains differed in endogenous viral superantigens (vSAGs), DNA of three D2×D recombinant inbred mouse lines was typed for endogenous mouse mammary tumor viruses using mouse mammary tumor virus long terminal repeat- and env gene-specific probes. The resistant D2×D mice were very similar to the susceptible parental strain D in their Mtv Southern blots, except for the lack of a single band corresponding to Mtv-7, the provirus coding for the strong DBA/2 superantigen Mls-1a. A backcross analysis revealed that Mtv-7-negative F2 mice were significantly more resistant than Mtv-7-positive F2 mice. When Mtv-7 was reintroduced into the resistant lines by crossing them with either CBA/J or BALB/D2.Mls-1a, the mice became again more tumor susceptible. Finally, we demonstrate the ability to transfer immunoresistance and graft-vs-leukemia reactivity from tumor-resistant to tumor-susceptible mice.
Collapse
Affiliation(s)
- Volker Schirrmacher
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Ulrich Beutner
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Marianna Bucur
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Victor Umansky
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Marian Rocha
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Paul von Hoegen
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
32
|
Reuss FU, Coffin JM. Mouse mammary tumor virus superantigen expression in B cells is regulated by a central enhancer within the pol gene. J Virol 1998; 72:6073-82. [PMID: 9621071 PMCID: PMC110413 DOI: 10.1128/jvi.72.7.6073-6082.1998] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Expression of mouse mammary tumor virus (MMTV)-encoded superantigens in B lymphocytes is required for viral transmission and pathogenesis. The mechanism of superantigen expression from the viral sag gene in B cells is largely unknown, due to problems with detection and quantification of these low-abundance proteins. We have established a sensitive superantigen-luciferase reporter assay to study the expression and regulation of the MMTV sag gene in B-cell lymphomas. The regulatory elements for retroviral gene expression are generally located in the 5' long terminal repeat (LTR) of the provirus. However, we found that neither promoters nor enhancers in the MMTV 5' LTR play a significant role in superantigen expression in these cells. Instead, the essential regulatory regions are located in the pol and env genes of MMTV. We report here that maximal sag expression in B-cell lines depends on an enhancer within the viral pol gene which can be localized to a minimal 183-bp region. Regulation of sag gene expression differs between B-cell lymphomas and pro-B cells, where an enhancer within the viral LTRs is involved. Thus, MMTV sag expression during B-cell development is achieved through the use of two separate enhancer elements.
Collapse
Affiliation(s)
- F U Reuss
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | |
Collapse
|
33
|
Taube R, Loya S, Avidan O, Perach M, Hizi A. Reverse transcriptase of mouse mammary tumour virus: expression in bacteria, purification and biochemical characterization. Biochem J 1998; 329 ( Pt 3):579-87. [PMID: 9445385 PMCID: PMC1219079 DOI: 10.1042/bj3290579] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have constructed a plasmid that induces in bacteria the synthesis of an enzymically active reverse transcriptase (RT) of mouse mammary tumour virus (MMTV), a retrovirus with a typical B-type morphology. The highest catalytic activity was detected only when 27 residues from the C-terminus of the protease were included in the N-terminus of the recombinant RT, after an extra deoxyadenosine was added between the pro and pol genes to overcome the -1 frameshift event (which occurs naturally in virus-infected cells). The recombinant protein with a six-histidine tag was purified to homogeneity by a two-column purification procedure, Ni2+ nitriloacetic acid/agarose followed by carboxymethyl-Sepharose chromatography. Unlike most RTs, the purified MMTV RT is enzymically active as a monomer even after binding a DNA substrate. Like all RTs studied, the recombinant MMTV RT possesses RNA-dependent and DNA-dependent DNA polymerase activities as well as RNase H activity, all of which show a preference for Mg2+ over Mn2+ ions. Other features of these enzymic activities, such as extension of DNA primers, processivity of DNA synthesis, pH dependence, steady-state kinetic constants, effects of Na+ or K+ ions and sensitivity to a thiol-specific reagent and to a zinc chelator, have been evaluated. The catalytic properties of MMTV RT were compared with those of the well-studied RT of HIV-1, the causative agent of AIDS. Interestingly, MMTV RT exhibits a high sensitivity to nucleoside triphosphate analogues (which are known to be potent inhibitors of HIV RTs and are being used as the major anti-AIDS drugs), as high as that of HIV-1 and HIV-2 RTs. Furthermore the recombinant MMTV RT shows a processivity of DNA synthesis higher than that of HIV-1 RT.
Collapse
Affiliation(s)
- R Taube
- Department of Cell Biology and Histology, Sackler School of Medicine, Tel Aviv University, Israel
| | | | | | | | | |
Collapse
|
34
|
Xu L, Wrona TJ, Dudley JP. Strain-specific expression of spliced MMTV RNAs containing the superantigen gene. Virology 1997; 236:54-65. [PMID: 9299617 DOI: 10.1006/viro.1997.8717] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The transmission of milk-borne or exogenous mouse mammary tumor virus (MMTV) requires infection of B cells in the gut-associated lymphoid tissue and expression of the superantigen (Sag) protein at the B-cell surface. Presentation of Sag at the B-cell surface is required for the transmission of MMTV to T cells and subsequent infection of the target mammary gland tissue. Because several different promoters have been reported for MMTV sag mRNA expression, we investigated whether the detection of spliced sag RNAs was dependent upon the cell type infected or the particular MMTV strain examined. In this study, we detected expression of spliced sag RNA from the standard promoter and from an internal U3 promoter in B-cell lines expressing endogenous Mtv-6 by RT-PCR, although expression from the standard promoter appeared to be at least 10-fold higher than that observed from the internal U3 promoter. Sag RNA originating from exogenous C3H MMTV was not observed from either of the U3 promoters in any cell type examined. However, spliced mRNAs containing the exogenous C3H MMTV, endogenous Mtv-8, or endogenous Mtv-17 sag genes could be detected from a previously described promoter in the envelope coding region regardless of the cell type infected. Because sag-specific RNAs can be initiated independently of the LTR promoters, there may be selection for independent control of MMTV sag and structural gene expression.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/biosynthesis
- Antigens, Viral/genetics
- B-Lymphocytes/immunology
- B-Lymphocytes/virology
- Base Sequence
- Consensus Sequence
- Exons
- Genes, Viral
- Genes, env
- Genes, pol
- Introns
- Lymphoma, B-Cell
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred Strains
- Molecular Sequence Data
- Polymerase Chain Reaction
- Promoter Regions, Genetic
- Proviruses/genetics
- Proviruses/physiology
- RNA Splicing
- RNA, Viral/biosynthesis
- Sequence Alignment
- Superantigens/biosynthesis
- Superantigens/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/virology
- Transcription, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- L Xu
- Department of Microbiology, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | |
Collapse
|
35
|
Lund AH, Duch M, Lovmand J, Jørgensen P, Pedersen FS. Complementation of a primer binding site-impaired murine leukemia virus-derived retroviral vector by a genetically engineered tRNA-like primer. J Virol 1997; 71:1191-5. [PMID: 8995641 PMCID: PMC191172 DOI: 10.1128/jvi.71.2.1191-1195.1997] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Reverse transcription of retroviral genomes is primed by a tRNA annealed to an 18-nucleotide primer binding site. Here, we present a primer complementation system to study molecular interaction of the replication machinery with the primer and primer binding site in vivo. Introduction of eight base substitutions into the primer binding site of a murine leukemia virus-based vector allowed efficient RNA encapsidation but resulted in severely reduced vector replication capacity. Replication was restored upon complementation with a synthetic gene designed to encode a complementary tRNA-like primer, but not with a noncomplementary tRNA-like molecule. The engineered primer was shown to be involved in both the initiation of first-strand synthesis and second-strand transfer. These results provide an in vivo demonstration that the retroviral replication machinery may recognize sequence complementarity rather than actual primer binding site and 3' primer sequences. Use of mutated primer binding site vectors replicating via engineered primers may add additional control features to retroviral gene transfer technology.
Collapse
Affiliation(s)
- A H Lund
- Department of Molecular and Structural Biology, University of Aarhus, Denmark
| | | | | | | | | |
Collapse
|
36
|
Mouse Mammary Tumor Virus: Immunological Interplays between Virus and Host **This article was accepted for publication on 1 October 1996. Adv Immunol 1997. [DOI: 10.1016/s0065-2776(08)60743-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
37
|
Nakamura N, Wajjwalku W, Nishimura H, Okubo H, Niimi N, Ando Y, Yoshikai Y. Nucleotide sequences of env and 3'LTROrf genes of endogenous mouse mammary tumor viruses encoding superantigen specific for TcrVbeta2. Immunogenetics 1996; 44:319-20. [PMID: 8753866 DOI: 10.1007/bf02602565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- N Nakamura
- Laboratory of Host Defense and Germfree Life, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Nagoya 466, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Wei WZ, Gill RF, Jones RF, Lichlyter D, Abastado JP. Induction of cytotoxic T lymphocytes to murine mammary tumor cells with a Kd-restricted immunogenic peptide. Int J Cancer 1996; 66:659-63. [PMID: 8647629 DOI: 10.1002/(sici)1097-0215(19960529)66:5<659::aid-ijc13>3.0.co;2-#] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A synthetic peptide E474 SFAVATTAL, derived from the sequence of mouse mammary tumor virus envelope protein, was previously shown to bind class I MHC Kd. Immunization of BALB/c mice with E474 in 50% incomplete Freund's adjuvant (IFA) followed by in vitro stimulation of immune cells with E474-coated antigen-presenting cells resulted in peptide-specific cytotoxic T lymphocytes (CTL). Furthermore, anti-E474 CTL lysed mammary tumor cell lines D2F2 and D2A1, derived from a spontaneous tumor that arose in BALB/c pre-neoplastic hyperplastic alveolar nodule (HAN) D2 line. Expression of Kd by D2A1 and D2F2 cells was verified by flow cytometry, and lysis of D2 tumor cells was blocked by monoclonal antibody 31-34-S, which interacted with the peptide-binding region of Kd, supporting the recognition of E474 in Kd by anti-E474 CTL. Immunization of BALB/c mice with E474 before D2F2 tumor challenge resulted in reduced tumor growth. Therefore, E474 is naturally processed and presented by these tumor cells and can induce anti-tumor immunity.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/pharmacology
- Cell Division/drug effects
- Histocompatibility Antigens Class I/biosynthesis
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/pharmacology
- Lymphocyte Activation/drug effects
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Tumor Virus, Mouse/chemistry
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Oligopeptides/biosynthesis
- Oligopeptides/immunology
- Oligopeptides/pharmacology
- Peptide Biosynthesis
- Peptides/immunology
- Peptides/pharmacology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Viral Envelope Proteins/biosynthesis
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/pharmacology
Collapse
Affiliation(s)
- W Z Wei
- Department of Immunology, Breast Cancer Program, Karmanos Cancer Institute, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
39
|
Shekels LL, Lyftogt C, Kieliszewski M, Filie JD, Kozak CA, Ho SB. Mouse gastric mucin: cloning and chromosomal localization. Biochem J 1995; 311 ( Pt 3):775-85. [PMID: 7487932 PMCID: PMC1136070 DOI: 10.1042/bj3110775] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mucins protect gastric epithelium by maintaining a favourable pH gradient and preventing autodigestion. The purpose of this study was to clone a mouse gastric mucin which would provide a foundation for analysis of mucin gene regulation. Mucin was purified from the glandular portion of gastric specimens and deglycosylated by HF solvolysis. Antibodies against native and deglycosylated mouse gastric mucin (MGM) were raised in chickens. Screening of a mouse stomach cDNA library with the anti-(deglycosylated MGM) antibody yielded partial clones containing a 48 bp tandem repeat and 768 bp of non-repetitive sequence. The 16-amino-acid tandem repeat has a consensus sequence of QTSSPNTGKTSTISTT with 25% serine and 38% threonine. The MGM tandem repeat sequence bears no similarity to previously identified mucins. The MGM non-repetitive region shares sequence similarity with human MUC5AC and, to a lesser extent, human MUC2 and rat intestinal mucin. Northern blot analysis reveals a polydisperse message beginning at 13.5 kb in mouse stomach with no expression in oesophagus, trachea, small intestine, large intestine, caecum, lung or kidney. Immunoreactivity of antibodies against deglycosylated MGM and against a synthetic MGM tandem repeat peptide was restricted to superficial mucous cells, antral glands and Brunner's glands in the pyloric-duodenal region. DNA analysis shows that MGM recognizes mouse and rat DNA but not hamster, rabbit or human DNA. The MGM gene maps to a site on mouse chromosome 7 homologous to the location of a human secretory mucin gene cluster on human chromosome 11p15. Due to sequence similarity and predominant expression in the stomach, the MGM gene may be considered a MUC5AC homologue and named Muc5ac.
Collapse
Affiliation(s)
- L L Shekels
- Department of Medicine, University of Minnesota, Minneapolis 55417, USA
| | | | | | | | | | | |
Collapse
|
40
|
Reuss FU, Coffin JM. Stimulation of mouse mammary tumor virus superantigen expression by an intragenic enhancer. Proc Natl Acad Sci U S A 1995; 92:9293-7. [PMID: 7568120 PMCID: PMC40971 DOI: 10.1073/pnas.92.20.9293] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The mechanisms regulating expression of mouse mammary tumor virus (MMTV)-encoded superantigens from the viral sag gene are largely unknown, due to problems with detection and quantification of these low-abundance proteins. To study the expression and regulation of the MMTV sag gene, we have developed a sensitive and quantitative reporter gene assay based on a recombinant superantigen-human placental alkaline phosphatase fusion protein. High sag-reporter expression in Ba/F3, an early B-lymphoid cell line, depends on enhancers in either of the viral long terminal repeats (LTRs) and is largely independent of promoters in the 5' LTR. The same enhancer region is also required for general expression of MMTV genes from the 5' LTR. The enhancer was mapped to a 548-bp fragment of the MMTV LTR lying within sag and shown to be sufficient to stimulate expression from a heterologous simian virus 40 promoter. No enhancer activity of the MMTV LTR was observed in XC sarcoma cells, which are permissive for MMTV. Our results demonstrate a major role for this enhancer in MMTV gene expression in early B-lymphoid cells.
Collapse
Affiliation(s)
- F U Reuss
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | |
Collapse
|
41
|
Xu W, Kozak CA, Desnick RJ. Uroporphyrinogen-III synthase: molecular cloning, nucleotide sequence, expression of a mouse full-length cDNA, and its localization on mouse chromosome 7. Genomics 1995; 26:556-62. [PMID: 7607680 DOI: 10.1016/0888-7543(95)80175-l] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Uroporphyrinogen-III synthase (URO-S; EC 4.2.1.75), the fourth enzyme in the heme biosynthetic pathway, is responsible for the conversion of hydroxymethylbilane to the cyclic tetrapyrrole, uroporphyrinogen III. The deficient activity of URO-S is the enzymatic defect in congenital erythropoietic porphyria (CEP), an autosomal recessive disorder. For the generation of a mouse model of CEP, the human URO-S cDNA was used to screen 2 x 10(6) recombinants from a mouse adult liver cDNA library. Ten positive clones were isolated, and dideoxy sequencing of the entire 1.6-kb insert of clone pmUROS-1 revealed 5' and 3' untranslated sequences of 144 and 623 bp, respectively, and an open reading frame of 798 bp encoding a 265-amino-acid polypeptide with a predicted molecular mass of 28,501 Da. The mouse and human coding sequences had 80.5 and 77.8% nucleotide and amino acid identity, respectively. The authenticity of the mouse cDNA was established by expression of the active monomeric enzyme in Escherichia coli. In addition, the analysis of two multilocus genetic crosses localized the mouse gene on chromosome 7, consistent with the mapping of the human gene to a position of conserved synteny on chromosome 10. The isolation, expression, and chromosomal mapping of this full-length cDNA should facilitate studies of the structure and organization of the mouse genomic sequence and the development of a mouse model of CEP for characterization of the disease pathogenesis and evaluation of gene therapy.
Collapse
Affiliation(s)
- W Xu
- Department of Human Genetics, Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | | | | |
Collapse
|
42
|
Cho K, Ferrick DA, Morris DW. Structure and biological activity of the subgenomic Mtv-6 endogenous provirus. Virology 1995; 206:395-402. [PMID: 7831795 DOI: 10.1016/s0042-6822(95)80055-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The Mtv-6 provirus has an incomplete genome, but retains a functional superantigen gene (sag) which directs the thymic deletion of CD4+ T cells expressing T cell receptors containing the V beta 3 or V beta 5 chains. To better understand the Mtv-6 superantigen, the structure and biological activity of the Mtv-6 provirus was analyzed. First, the complete nucleotide sequence was determined, and the mutation producing the subgenomic provirus was identified. Second, the nucleotide sequence of the 5' end of the sag gene transcript (including the splice junction) was determined by sequence analysis of a cDNA clone. Third, the superantigen activity of Mtv-6 was analyzed in mice carrying the Mtv-6 provirus isolated by selective breeding on a genetic background free of endogenous and exogenous mouse mammary tumor virus (MMTV). These studies demonstrate that (i) the Mtv-6 provirus contains a 6.2-kb deletion between two 12-bp direct repeats encompassing the central portion of the provirus but not affecting sag gene splicing or translation, (ii) the sag gene transcript has the structure predicted from previous S1 nuclease mapping studies, and (iii) the Mtv-6 superantigen can direct thymic deletion of target V beta 3+ and V beta 5+ T cells in the absence of gene products from full-length MMTV proviruses.
Collapse
Affiliation(s)
- K Cho
- Department of Medical Pathology, University of California at Davis 95616
| | | | | |
Collapse
|
43
|
Wajjwalku W, Ando Y, Niimi N, Yoshikai Y. A novel exogenous mammary tumor virus encoding MHC class II H2E-independent superantigen specific for Tcr-V beta 14. Immunogenetics 1995; 41:156-8. [PMID: 7806290 DOI: 10.1007/bf00182330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Base Sequence
- CD4-Positive T-Lymphocytes/immunology
- Histocompatibility Antigens Class II/immunology
- Lymph Nodes/cytology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Sequence Homology, Amino Acid
- Superantigens/genetics
- Superantigens/immunology
Collapse
Affiliation(s)
- W Wajjwalku
- Department of Pathology, Faculty of Veterinary Medicine, Kasetstart University, Nakhonpathom, Thailand
| | | | | | | |
Collapse
|
44
|
Kozak CA, Gatignol A, Graham K, Jeang KT, McBride OW. Genetic mapping in human and mouse of the locus encoding TRBP, a protein that binds the TAR region of the human immunodeficiency virus (HIV-1). Genomics 1995; 25:66-72. [PMID: 7774957 DOI: 10.1016/0888-7543(95)80110-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Productive infection with HIV-1, the virus responsible for AIDS, requires the involvement of host cell factors for completion of the replicative cycle, but the identification of these factors and elucidation of their specific functions has been difficult. A human cDNA, TRBP, was recently cloned and characterized as a positive regulator of gene expression that binds to the TAR region of the HIV-1 genome. Here we demonstrate that this factor is encoded by a gene, TARBP2, that maps to human chromosome 12 and mouse chromosome 15, and we also identify and map one human pseudogene (TARBP2P) and two mouse TRBP-related sequences (Tarbp2-rs1, Tarbp2-rs2). The map location of the expressed gene identifies it as a candidate for the previously identified factor encoded on human chromosome 12 that has been shown to be important for expression of HIV-1 genes. Western blotting indicates that despite high sequence conservation in human and mouse, the TARBP2 protein differs in apparent size in primate and rodent cells.
Collapse
Affiliation(s)
- C A Kozak
- Laboratory of Molecular Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
45
|
Szurek PF, Brooks BR. Development of physical forms of unintegrated retroviral DNA in mouse spinal cord tissue during ts1-induced spongiform encephalomyelopathy: elevated levels of a novel single-stranded form in paralyzed mice. J Virol 1995; 69:348-56. [PMID: 7983729 PMCID: PMC188582 DOI: 10.1128/jvi.69.1.348-356.1995] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
ts1 is a murine leukemia virus that causes rapidly evolving hindlimb paralysis in susceptible strains of mice. Following perinatal infection, three physical forms of unintegrated viral DNA were detected in the spinal cord by Southern blot hybridization. Linear and supercoiled closed-circle viral double-stranded DNAs were detected in both the central nervous system and non-central nervous system tissues. An elevated level of a novel minus-sense single-stranded form of viral DNA, which had a very high mobility in agarose gels, was correlated with the onset of symptoms of paralysis. As the severity of paralysis progressed, the level of this single-stranded form increased rapidly, with the highest level in the spinal cords of moribund mice. Since the virulence of a number of cytopathic retroviruses has been associated with the presence of increased amounts of unintegrated viral DNA in the tissues of the infected hosts, this novel form of highly mobile unintegrated single-stranded DNA may have a role in the neuropathogenesis of ts1.
Collapse
Affiliation(s)
- P F Szurek
- Neurology Service, William S. Middleton Memorial Veterans Affairs Hospital
| | | |
Collapse
|
46
|
Gill RF, Abastado JP, Wei WZ. Systematic identification of H-2 Kd binding peptides and induction of peptide specific CTL. J Immunol Methods 1994; 176:245-53. [PMID: 7983382 DOI: 10.1016/0022-1759(94)90318-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Most peptides with putative MHC I restricted sequence motifs do not bind to the corresponding MHC I nor induce cytolytic T cells. There exist additional constraints which limit peptide binding and immunogenicity. To identify immunogenic peptides in novel protein sequences, it will be necessary to first evaluate peptide binding to MHC I. In this study, a soluble single chain fusion protein SC-Kd was used to evaluate potential Kd binding peptides from the sequences of mouse mammary tumor virus gag and env proteins. A total of 27 peptides were identified which displayed the reported Kd restricted motif. Of the 27 peptides, six demonstrated strong to moderate binding to SC-Kd. The strongest binding peptides expressed tyrosine or phenylalanine at position 2 and leucine at the C-terminus. The capability of MMTV peptides to induce CTL corresponds to their SC-Kd binding activity. Of the six peptides that demonstrated moderate to strong binding, five induced CTL in BALB/c mice. These peptides induced CTL after 1-3 in vivo immunizations followed by 5 day in vitro stimulation. Furthermore, a single in vitro stimulation of naive lymphocytes with strong-binding G425 was sufficient to induce significant CTL activity. Weak or non-binding peptides did not induce CTL. Therefore, peptide binding to SC-Kd is a predictive indicator of CTL inducing activity.
Collapse
Affiliation(s)
- R F Gill
- Department of Immunology, Michigan Cancer Foundation, Detroit 48201
| | | | | |
Collapse
|
47
|
Xu L, Haga S, Imai S, Sarkar NH. Cloning in a plasmid of an MMTV from a wild Chinese mouse: sequencing of the viral LTR. Virus Res 1994; 33:167-78. [PMID: 7975881 DOI: 10.1016/0168-1702(94)90053-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Plasmid subcloning by conventional techniques of full length exogenous mouse mammary viruses (MMTV) has not been realized because of the involvement of host-mediated structural changes in the viral gag gene. To circumvent this problem, an alternative subcloning method, excision of phagemid (pBluescript SK) from lambda ZAP II, was successfully used to subclone a novel exogenous MMTV (JYG-MMTV) provirus fragment containing an intact gag gene. Sequence analysis revealed that the LTR of this virus is significantly different from the LTR of C3H-MMTV in the U3 region.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Animals, Wild
- Base Sequence
- Cloning, Molecular
- DNA, Viral/genetics
- Female
- Genes, gag
- Male
- Mammary Neoplasms, Experimental/virology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred C3H
- Molecular Sequence Data
- Plasmids/genetics
- Proviruses/genetics
- Repetitive Sequences, Nucleic Acid
- Restriction Mapping
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
Collapse
Affiliation(s)
- L Xu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta 30912
| | | | | | | |
Collapse
|
48
|
Roger T, Boudaly S, Seman M. Negative segregation of Mtv loci in H-2E+ mice selected for high antibody response. Immunogenetics 1994; 40:123-8. [PMID: 8026861 DOI: 10.1007/bf00188175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Endogenous mouse mammary tumor proviruses (Mtvs) encode superantigens (Sags), which can delete T lymphocytes expressing particular Tcrb-V genes when associated with the H-2E molecule. In the present work, distribution of Mtvs was investigated in six independent pairs of mouse lines genetically selected for high (H) or low (L) Ab production to specific T-cell-dependent Ags. These experiments were performed to evaluate the role of Mtv-encoded Sags in the determination of H and L phenotypes. No systematic difference is observed in Mtv segregation between H-2E- H and L mouse lines. However, a clear differential segregation of Mtv loci is observed between H-2E+ H and L mice from three independent selections. The number of endogenous Mtvs in L lines is close to that found in laboratory mice. In contrast, Mtv loci-encoding Sags are almost absent in H animals. Although Sags profoundly affect the available Tcrb-V repertoire in H-2E+ L mice, it seems unlikely that Mtvs account for the L phenotype which can be achieved independently of Mtv segregation in H-2E- lines. More importantly, the results suggest that negative segregation of Mtv-encoding Sags contribute to determine the super-responder phenotype of H lines.
Collapse
Affiliation(s)
- T Roger
- Laboratorie d'Immunodifférenciation, Hall de Biotechnologies, Université Denis Diderot (Paris 7), France
| | | | | |
Collapse
|
49
|
Beutner U, Kraus E, Kitamura D, Rajewsky K, Huber BT. B cells are essential for murine mammary tumor virus transmission, but not for presentation of endogenous superantigens. J Exp Med 1994; 179:1457-66. [PMID: 8163931 PMCID: PMC2191484 DOI: 10.1084/jem.179.5.1457] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Murine mammary tumor viruses (MMTVs) are retroviruses that encode superantigens capable of stimulating T cells via superantigen-reactive T cell receptor V beta chains. MMTVs are transmitted to the suckling offspring through milk. Here we show that B cell-deficient mice foster nursed by virus-secreting mice do not transfer infectious MMTVs to their offspring. No MMTV proviruses could be detected in the spleen and mammary tissue of these mice, and no deletion of MMTV superantigen-reactive T cells occurred. By contrast, T cell deletion and positive selection due to endogenous MMTV superantigens occurred in B cell-deficient mice. We conclude that B cells are essential for the completion of the viral life cycle in vivo, but that endogenous MMTV superantigens can be presented by cell types other than B cells.
Collapse
Affiliation(s)
- U Beutner
- Program of Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | | | | | | | | |
Collapse
|
50
|
Niimi N, Wajjwalku W, Ando Y, Tomida S, Takeuchi M, Ueda M, Kaneda T, Yoshikai Y. Delay in expression of a mammary tumor provirus is responsible for defective clonal deletion during postnatal period. Eur J Immunol 1994; 24:488-91. [PMID: 8299700 DOI: 10.1002/eji.1830240235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A gene-encoding ligand for deletion of T cells bearing TcRV beta 6 and V beta 8.1 cosegregates a new mammary tumor provirus locus, Mtv-50 in NC mice. The sequence of the open reading frame (ORF) in the 3' long terminal repeat (LTR) of Mtv-50 was strikingly similar to those of Mtv-7, Mtv-43 and exogenous mouse mammary tumor virus (SW) with properties of minor lymphocyte stimulating antigen 1a. Consistent with previous reports, clonal deletion of mature thymocytes bearing TcRV beta 6 was defective during the early postnatal period of mice carrying Mtv-50. Appreciable levels of mRNA corresponding to common Mtv ORF and Mtv-6 ORF were expressed in the neonatal thymus, while little, if any, mRNA corresponding to Mtv-50 ORF was detected in the thymus at the early postnatal stage. Delay in expression of Mtv-50 ORF during the postnatal period may be responsible for the failure of clonal deletion of V beta 6-T cells in the early postnatal life of mice carrying Mtv-50.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Animals, Newborn/immunology
- Animals, Newborn/microbiology
- Base Sequence
- DNA Primers/chemistry
- Gene Expression Regulation, Viral
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred Strains
- Minor Lymphocyte Stimulatory Antigens/genetics
- Molecular Sequence Data
- RNA, Viral/analysis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Repetitive Sequences, Nucleic Acid
- Superantigens/immunology
- Thymus Gland/cytology
Collapse
Affiliation(s)
- N Niimi
- Department of Oral Surgery, Nagoya University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|