Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Nature ; 488(7411): 349-52, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22895340

ABSTRACT

In the cores of some clusters of galaxies the hot intracluster plasma is dense enough that it should cool radiatively in the cluster's lifetime, leading to continuous 'cooling flows' of gas sinking towards the cluster centre, yet no such cooling flow has been observed. The low observed star-formation rates and cool gas masses for these 'cool-core' clusters suggest that much of the cooling must be offset by feedback to prevent the formation of a runaway cooling flow. Here we report X-ray, optical and infrared observations of the galaxy cluster SPT-CLJ2344-4243 (ref. 11) at redshift z = 0.596. These observations reveal an exceptionally luminous (8.2 × 10(45) erg s(-1)) galaxy cluster that hosts an extremely strong cooling flow (around 3,820 solar masses a year). Further, the central galaxy in this cluster appears to be experiencing a massive starburst (formation of around 740 solar masses a year), which suggests that the feedback source responsible for preventing runaway cooling in nearby cool-core clusters may not yet be fully established in SPT-CLJ2344-4243. This large star-formation rate implies that a significant fraction of the stars in the central galaxy of this cluster may form through accretion of the intracluster medium, rather than (as is currently thought) assembling entirely via mergers.

2.
Zoonoses Public Health ; 56(6-7): 357-69, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19486320

ABSTRACT

The observed patterns and variations in the ecology, epidemiology, distribution and prevalence of the West Nile Virus (WNV) in different areas of the Western Hemisphere make this pathogen of particular importance as a model for understanding the potential risk factors associated with emerging pathogens worldwide, particularly those involving zoonotic pathogens whose epidemiology involves the potential for vertical transmission in arthropod vector species, and horizontal and vertical transmission within and among vertebrate host species. Record numbers of human WNV cases were recorded in Canada during 2007, with >50% more cases than documented in any previous year. Although overall numbers of human infections recorded in the United States were not exceptionally high during 2007 relative to epidemic levels reported in 2002 and 2003, the state of Oklahoma reported that the highest-ever number of human WNV cases and the numbers of human cases recorded in Canada were 50% higher than previous record levels recorded in 2003. The record and near-record numbers of human WNV infections recorded in several regions of North America during 2007 have important implications for the future management and surveillance of WNV vectors and reservoirs in North America. The spatiotemporal distribution of WNV infections in humans and animals recorded during 2007 in North America and South America have important implications for the surveillance and management of public health threats from WNV in the Western Hemisphere. Serological surveys conducted in areas of intense WNV transmission in the United States have reported low prevalence of antibodies to WNV in human s populations, indicating that additional epidemic outbreaks of human disease from WNV can be expected in the future.


Subject(s)
Communicable Diseases, Emerging/veterinary , Global Health , West Nile Fever/transmission , West Nile Fever/veterinary , Zoonoses , Animals , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/transmission , Communicable Diseases, Emerging/virology , Disease Outbreaks/veterinary , Humans , Sentinel Surveillance , West Nile Fever/epidemiology , West Nile virus
3.
Euro Surveill ; 14(18)2009 May 07.
Article in English | MEDLINE | ID: mdl-19422779

ABSTRACT

The age-specific infection and death profiles among confirmed human cases of influenza A(H5N1) infection in Egypt differ markedly from those recorded in other countries. The case fatality rate among human H5N1 cases in Egypt is 34%, versus an average of 66% in other countries. In Egypt, children younger than 10 years comprise 48% of reported cases, nearly twice the global average of approximately 25%, and no H5N1 fatalities have been confirmed among individuals in this age group as of 23 April 2009. Females outnumber males among confirmed H5N1 cases by a factor of nearly 2:1, and 90% of reported fatalities in Egypt have been females. The evident age and sex biases in morbidity and mortality among H5N1 cases in Egypt are phenomena that warrant further investigation and analysis.


Subject(s)
Creutzfeldt-Jakob Syndrome/epidemiology , Disease Outbreaks/statistics & numerical data , Influenza A Virus, H5N1 Subtype , Influenza, Human/mortality , Population Surveillance , Risk Assessment/methods , Europe/epidemiology , Humans , Incidence , Risk Factors , Survival Analysis , Survival Rate
4.
Leukemia ; 16(6): 1086-98, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040439

ABSTRACT

Overexpression of the cellular oncogene c-Myc frequently occurs during induction of leukemias and lymphomas in many species. Retroviruses have enhanced our understanding of the role of c-Myc in such tumors. Leukemias and lymphomas induced by retroviruses activate c-Myc by: (1) use of virally specified proteins that increase c-Myc transcription, (2) transduction and modification of c-Myc to generate a virally encoded form of the gene, v-Myc, and (3) proviral integration in or near c-Myc. Proviral integrations elevate transcription by insertion of retroviral enhancers found in the long terminal repeat (LTR). Studies of the LTR enhancer elements from these retroviruses have revealed the importance of these elements for c-Mycactivation in several cell types. Retroviruses also have been used to identify genes that collaborate with c-Myc during development and progression of leukemias and lymphomas. In these experiments, animals that are transgenic for c-Mycoverexpression (often in combination with the overexpression or deletion of known proto-oncogenes) have been infected with retroviruses that then insertionally activate novel co-operating cellular genes. The retrovirus then acts as a molecular 'tag' for cloning of these genes. This review covers several aspects of c-Myc involvement in retrovirally induced leukemias and lymphomas.


Subject(s)
Leukemia/virology , Lymphoma/virology , Proto-Oncogene Proteins c-myc/physiology , Retroviridae/genetics , Animals , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Leukemia/genetics , Leukemia/metabolism , Lymphoma/genetics , Lymphoma/metabolism , Oncogenes , Proto-Oncogene Proteins c-myc/genetics , Terminal Repeat Sequences , Virus Integration
5.
Rev Sci Tech ; 21(1): 125-37, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11974624

ABSTRACT

Diseases and biological toxins have been used as weapons of war throughout recorded history, from Biblical times through to the present day. Bioweapon uses have historically been directed primarily, although not exclusively, against human populations. Specialised technicians and state-of-the-art research facilities are no longer necessary for the production or deployment of many known bioweapon agents and commercially available technologies now permit the large-scale production of bioweapon agents in small-scale facilities at relatively low cost. Failures in the detection and containment of bioweapon and emerging disease outbreaks among populations of wildlife and indigenous peoples in developing countries could result in severe erosion of genetic diversity in local and regional populations of both wild and domestic animals, the extinction of endangered species and the extirpation of indigenous peoples and their cultures. Our ability to understand and control the spread of diseases within and among human and animal populations is increasing but is still insufficient to counter the threats presented by existing bioweapon diseases and the growing number of highly pathogenic emergent infections. Interdisciplinary and international efforts to increase the monitoring, surveillance, identification and reporting of disease agents and to better understand the potential dynamics of disease transmission within human and animal populations in both industrialised and developing country settings will greatly enhance our ability to combat the effects of bioweapons and emerging diseases on biological communities and biodiversity.


Subject(s)
Biological Warfare , Bioterrorism , Ecosystem , Agriculture/economics , Agriculture/trends , Animals , Animals, Domestic , Animals, Wild , Biological Warfare/methods , Biological Warfare/prevention & control , Biotechnology/trends , Bioterrorism/prevention & control , Communicable Diseases/epidemiology , Communicable Diseases/veterinary , Communicable Diseases, Emerging/prevention & control , Disease Outbreaks/economics , Disease Outbreaks/veterinary , Genetic Variation , Humans
6.
J Virol ; 75(5): 2174-84, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11160721

ABSTRACT

Type B leukemogenic virus (TBLV) induces rapidly appearing T-cell tumors in mice. TBLV is highly related to mouse mammary tumor virus (MMTV) except that TBLV long terminal repeats (LTRs) have a deletion of negative regulatory elements and a triplication of sequences flanking the deletion. To determine if the LTR triplication represents a viral enhancer element, we inserted the triplication upstream and downstream in either orientation relative to the thymidine kinase promoter linked to the luciferase gene. These experiments showed that upregulation of reporter gene activity by the TBLV triplication was relatively orientation independent, consistent with the activity of eukaryotic enhancer elements. TBLV enhancer activity was observed in T-cell lines but not in fibroblasts, B cells, or mammary cells, suggesting that enhancer function is cell type dependent. To analyze the transcription factor binding sites that are important for TBLV enhancer function, we prepared substitution mutations in a reconstituted C3H MMTV LTR that recapitulates the deletion observed in the TBLV LTR. Transient transfections showed that a single mutation (556M) decreased TBLV enhancer activity at least 20-fold in two different T-cell lines. This mutation greatly diminished AML-1 (recently renamed RUNX1) binding in gel shift assays with a mutant oligonucleotide, whereas AML-1 binding to a wild-type TBLV oligomer was specific, as judged by competition and supershift experiments. The 556 mutation also reduced TBLV enhancer binding of two other protein complexes, called NF-A and NF-B, that did not appear to be related to c-Myb or Ets. AML-1 overexpression in a mammary cell line enhanced expression from the TBLV LTR approximately 30-fold. These data suggest that binding of AML-1 to the TBLV enhancer, likely in combination with other factors, is necessary for optimal enhancer function.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/physiology , Proto-Oncogene Proteins , Retroviridae/genetics , T-Lymphocytes/virology , Terminal Repeat Sequences/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Core Binding Factor Alpha 2 Subunit , Enhancer Elements, Genetic/genetics , Genes, Reporter , Humans , Jurkat Cells , Luciferases/genetics , Luciferases/metabolism , Mice , Molecular Sequence Data , Mutation , Plasmids/genetics , Rats , Retroviridae/physiology , T-Lymphocytes/physiology , Terminal Repeat Sequences/physiology , Transcription, Genetic , Transfection
7.
J Virol ; 74(20): 9431-40, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000212

ABSTRACT

Mouse mammary tumor virus (MMTV) encodes a superantigen (Sag) that is required for efficient milk-borne transmission of virus from mothers to offspring. The mRNA used for Sag expression is controversial, and at least four different promoters (two in the long terminal repeat and two in the envelope gene) for sag mRNA have been reported. To determine which RNA is responsible for Sag function during milk-borne MMTV transmission, we mutated a splice donor site unique to a spliced sag RNA from the 5' envelope promoter. The splice donor mutation in an infectious provirus was transfected into XC cells and injected into BALB/c mice. Mice injected with wild-type provirus showed Sag activity by the deletion of Sag-specific T cells and induction of mammary tumors in 100% of injected animals. However, mice injected with the splice donor mutant gave sporadic and delayed T-cell deletion and a low percentage of mammary tumors with a long latency, suggesting that the resulting tumors were due to the generation of recombinants with endogenous MMTVs. Third-litter offspring of mice injected with wild-type provirus showed Sag-specific T-cell deletion and developed mammary tumors with kinetics similar to those for mice infected by nursing on MMTV-infected mothers, whereas the third-litter offspring of the splice donor mutant-injected mice did not. One of the fifth-litter progeny of splice donor mutant-injected mice showed C3H Sag activity and had recombinants that repaired the splice donor mutation, thus confirming the necessity for the splice donor site for Sag function. These experiments are the first to show that the spliced sag mRNA from the 5' envelope promoter is required for efficient milk-borne transmission of C3H MMTV.


Subject(s)
Genes, env , Mammary Tumor Virus, Mouse/immunology , Superantigens/genetics , Animals , Base Sequence , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Promoter Regions, Genetic , RNA Splicing , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Superantigens/physiology , Terminal Repeat Sequences
8.
J Virol ; 74(14): 6348-57, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10864645

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.


Subject(s)
Homeodomain Proteins/metabolism , Mammary Glands, Animal/metabolism , Mammary Tumor Virus, Mouse/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cells, Cultured , DNA Footprinting , DNA, Viral/genetics , DNA, Viral/physiology , Deoxyribonuclease I , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/virology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred BALB C , Mutagenesis , Nuclear Proteins/genetics , Protein Binding , Repressor Proteins/genetics , Terminal Repeat Sequences
9.
J Virol ; 74(5): 2466-71, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10666282

ABSTRACT

Type B leukemogenic virus (TBLV) induces rapidly appearing T-cell leukemias. TBLV insertions near the c-myc gene were detectable in 2 of 30 tumors tested, whereas 80% of the tumors showed c-myc overexpression. TBLV insertions on chromosome 15 (including a newly identified locus, Pad7) may cause c-myc overexpression by cis-acting effects at a distance.


Subject(s)
Betaretrovirus/genetics , Genes, myc/genetics , Lymphoma, T-Cell/genetics , Animals , Chromosome Mapping , Lymphoma, T-Cell/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Virus Integration/genetics
10.
J Virol ; 73(8): 6634-45, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400761

ABSTRACT

Mouse mammary tumor virus (MMTV) encodes a superantigen (Sag) that is expressed at the surface of antigen-presenting cells in conjunction with major histocompatibility complex (MHC) type II molecules. The Sag-MHC complex is recognized by entire subsets of T cells, leading to cytokine release and amplification of infected B and T cells that carry milk-borne MMTV to the mammary gland. Expression of Sag proteins from endogenous MMTV proviruses carried in the mouse germ line usually results in the deletion of self-reactive T cells during negative selection in the thymus and the elimination of T cells required for infection by specific milk-borne MMTVs. However, other endogenous MMTVs are unable to eliminate Sag-reactive T cells in newborn mice and cause partial loss of reactive T cells in adults. To investigate the kinetics of Sag-reactive T-cell deletion, backcross mice that contain single or multiple MMTVs were screened by a novel PCR assay designed to distinguish among highly related MMTV strains. Mice that contained Mtv-17 alone showed slow kinetics of reactive T-cell loss that involved the CD4(+), but not the CD8(+), subset. Deletion of CD4(+) or CD8(+) T cells reactive with Mtv-17 Sag was not detected in thymocytes. Slow kinetics of peripheral T-cell deletion by Mtv-17 Sag also was accompanied by failure to detect Mtv-17 sag-specific mRNA in the thymus, despite detectable expression in other tissues, such as spleen. Together, these data suggest that Mtv-17 Sag causes peripheral, rather than intrathymic, deletion of T cells. Interestingly, the Mtv-8 provirus caused partial deletion of CD4(+)Vbeta12(+) cells in the thymus, but other T-cell subsets appeared to be deleted only in the periphery. Our data have important implications for the level of antigen expression required for elimination of self-reactive T cells. Moreover, these experiments suggest that mice expressing endogenous MMTVs that lead to slow kinetics of T-cell deletion will be susceptible to infection by milk-borne MMTVs with the same Sag specificity.


Subject(s)
Antigens, Viral/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Depletion , Mammary Tumor Virus, Mouse/immunology , RNA, Viral , Superantigens/biosynthesis , Thymus Gland/immunology , Animals , Antigens, Viral/genetics , Female , Gene Expression , Kinetics , Male , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred BALB C , Proviruses , RNA, Messenger , Superantigens/genetics , Thymus Gland/cytology
11.
Mol Cell Biol ; 19(7): 4918-26, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373541

ABSTRACT

Homeoproteins are known to participate in development and cell type specification. The homeoproteins CCAAT displacement protein (CDP) and special AT-rich sequence binding protein 1 (SATB1) have been shown to bind to nuclear matrix-associated regions and to act as repressors of many cellular genes. Moreover, binding of SATB1 to the mouse mammary tumor virus (MMTV) promoter region dramatically affects the tissue-specific transcription of this retrovirus. Because protein-protein interactions are a common means of regulating homeoprotein function, we tested whether SATB1 and CDP interact in vivo and in vitro. SATB1 interacted with CDP through its DNA-binding domain, as demonstrated by glutathione S-transferase (GST) pull-down assays. GST pull-down assays also showed that CDP associated with SATB1 through three of its four DNA-binding domains (CR1, CR2, and the homeodomain). SATB1-specific antisera, but not preimmune sera, precipitated CDP from nuclear extracts, and CDP-specific antisera precipitated SATB1 from the same extracts. Far-Western blotting detected interaction of SATB1 and CDP in several different tissue extracts. Association of purified SATB1 and CDP in vitro resulted in the inability of each protein to bind to DNA in gel retardation assays. CDP overexpression in cultured T cells led to a loss of detectable SATB1 binding to the MMTV promoter region, as measured by gel shift experiments. CDP overexpression also elevated MMTV long terminal repeat reporter gene activity in transient-transfection assays, a result consistent with neutralization of the SATB1 repressor function in T cells. SATB1 is very abundant in certain tissues, particularly thymus, whereas CDP is relatively ubiquitous, except in certain terminally differentiated cell types. Because of the tissue and cell type distribution of SATB1 and CDP, we propose that the SATB1-to-CDP ratio in different tissues is a novel mechanism for homeoproteins to control gene expression and differentiation in mammals.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Matrix Attachment Region Binding Proteins , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Western , Cell Line , DNA/metabolism , DNA-Binding Proteins/genetics , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Homeodomain Proteins/genetics , Humans , Jurkat Cells , Mice , Mink , Nuclear Proteins/genetics , Precipitin Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors , Transfection
12.
J Immunol ; 161(5): 2375-82, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9725233

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, V beta 14+ 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 (mu-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.


Subject(s)
B-Lymphocytes/immunology , Mammary Glands, Animal/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse/immunology , Retroviridae Infections/immunology , T-Lymphocytes/immunology , Tumor Virus Infections/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , B-Lymphocytes/virology , Female , Gene Amplification/immunology , Mammary Glands, Animal/virology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pedigree , Retroviridae Infections/genetics , Retroviridae Infections/transmission , Superantigens/genetics , Superantigens/immunology , T-Lymphocytes/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/transmission
13.
J Virol ; 72(6): 4746-55, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9573239

ABSTRACT

The mouse mammary tumor virus (MMTV) encodes within the U3 region of the long terminal repeat (LTR) a protein known as the superantigen (Sag). Sag is needed for the efficient transmission of milk-borne virus from the gut to target tissue in the mammary gland. MMTV-infected B cells in the gut express Sag as a type II transmembrane protein that is recognized by the variable region of particular beta chains (Vbeta) of the T-cell receptor (TCR) on the surface of T cells. Recognition of Sag by particular TCRs results in T-cell stimulation, release of cytokines, and amplification of MMTV infection in lymphoid cells that are needed for infection of adolescent mammary tissue. Because the C-terminal 30 to 40 amino acids of Sag are variable and correlate with recognition of particular TCR Vbeta chains, we prepared a series of C-terminal Sag mutations that were introduced into a cloned infectious MMTV provirus. Virus-producing XC rat cells were used for injection of susceptible BALB/c mice, and these mice were monitored for functional Sag activity by the deletion of C3H MMTV Sag-reactive (CD4+ Vbeta14+) T cells. Injected mice also were analyzed for mutant infection and tumor formation in mammary glands as well as milk-borne transmission of MMTV to offspring. Most mutations abrogated Sag function, although one mutation (HPA242) that changed the negative charge of the extreme C terminus to a positive charge created a weaker Sag that slowed the kinetics of Sag-mediated T-cell deletion. Despite the lack of Sag activity, many of the sag mutant viruses were capable of sporadic infections of the mammary glands of injected mice but not of offspring mice, indicating that functional Sag increases the probability of milk-borne MMTV infection. Furthermore, although most viruses encoding nonfunctional Sags were unable to cause mammary tumors, tumors were induced by such viruses carrying mutations in a negative regulatory element that overlaps the sag gene within the LTR, suggesting that loss of Sag function may be compensated, at least partially, by loss of transcriptional suppression in certain tissues. Together these results confirm the importance of Sag for efficient milk-borne transmission and indicate that the entire C-terminal region is needed for complete Sag function.


Subject(s)
DNA, Viral/genetics , Mammary Tumor Virus, Mouse/genetics , Retroviridae Infections/virology , Superantigens/genetics , Tumor Virus Infections/virology , Amino Acid Sequence , Animals , DNA Mutational Analysis , Female , Genes, Viral , Mammary Tumor Virus, Mouse/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Mutation , Rats , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Retroviridae Infections/immunology , Sequence Alignment , Superantigens/immunology , Tumor Virus Infections/immunology
15.
Mol Cell Biol ; 17(9): 5275-87, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9271405

ABSTRACT

The nuclear matrix has been implicated in several cellular processes, including DNA replication, transcription, and RNA processing. In particular, transcriptional regulation is believed to be accomplished by binding of chromatin loops to the nuclear matrix and by the concentration of specific transcription factors near these matrix attachment regions (MARs). A number of MAR-binding proteins have been identified, but few have been directly linked to tissue-specific transcription. Recently, we have identified two cellular protein complexes (NBP and UBP) that bind to a region of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR) previously shown to contain at least two negative regulatory elements (NREs) termed the promoter-proximal and promoter-distal NREs. These NREs are absent from MMTV strains that cause T-cell lymphomas instead of mammary carcinomas. We show here that NBP binds to a 22-bp sequence containing an imperfect inverted repeat in the promoter-proximal NRE. Previous data showed that a mutation (p924) within the inverted repeat elevated basal transcription from the MMTV promoter and destabilized the binding of NBP, but not UBP, to the proximal NRE. By using conventional and affinity methods to purify NBP from rat thymic nuclear extracts, we obtained a single major protein of 115 kDa that was identified by protease digestion and partial sequencing analysis as the nuclear matrix-binding protein special AT-rich sequence-binding protein 1 (SATB1). Antibody ablation, distamycin inhibition of binding, renaturation and competition experiments, and tissue distribution data all confirmed that the NBP complex contained SATB1. Similar types of experiments were used to show that the UBP complex contained the homeodomain protein Cux/CDP that binds the MAR of the intronic heavy-chain immunoglobulin enhancer. By using the p924 mutation within the MMTV LTR upstream of the chloramphenicol acetyltransferase gene, we generated two strains of transgenic mice that had a dramatic elevation of reporter gene expression in lymphoid tissues compared with reporter gene expression in mice expressing wild-type LTR constructs. Thus, the 924 mutation in the SATB1-binding site dramatically elevated MMTV transcription in lymphoid tissues. These results and the ability of the proximal NRE in the MMTV LTR to bind to the nuclear matrix clearly demonstrate the role of MAR-binding proteins in tissue-specific gene regulation and in MMTV-induced oncogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Matrix Attachment Region Binding Proteins , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Cell Line , Distamycins/pharmacology , Female , Gene Expression , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , Male , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/metabolism , Rats , Repressor Proteins/metabolism , Transcription Factors
16.
Virology ; 236(1): 54-65, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9299617

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.


Subject(s)
Mammary Tumor Virus, Mouse/genetics , RNA Splicing , RNA, Viral/biosynthesis , Superantigens/biosynthesis , Transcription, Genetic , 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 , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred Strains , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Proviruses/genetics , Proviruses/physiology , Sequence Alignment , Superantigens/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Cells, Cultured
18.
J Virol ; 70(2): 1246-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8551589

ABSTRACT

C57BL/6 mice are resistant to C3H mouse mammary tumor virus (MMTV)-induced mammary tumorigenesis and lack major histocompatibility complex class II I-E molecules that are essential for presentation of C3H superantigen to T cells. T cells are needed for transmission of milk-borne MMTV from the gut to the mammary gland. In this report, we show that infectious C3H MMTV is produced by C57BL/6 mice that nurse on C3H mothers but that virus production in the mammary gland is delayed compared with that in I-E+ mouse strains.


Subject(s)
Histocompatibility Antigens Class II/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse/immunology , Retroviridae Infections/transmission , Tumor Virus Infections/transmission , Animals , Female , Histocompatibility Antigens Class II/genetics , Immunity, Innate/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/immunology , Retroviridae Infections/immunology , Tumor Virus Infections/immunology
19.
Virology ; 215(2): 113-23, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8560758

ABSTRACT

The mouse mammary tumor virus (MMTV) superantigen (Sag) protein is involved in the transmission of milk-borne MMTV from virus-infected milk in the gut to the target mammary gland tissue. Using an RT-PCR assay for in vivo MMTV infection, BALB/c or C3H mice nursed on C3H MMTV-infected mothers showed sag mRNA expression in intestine, spleen, and thymus as early as 1 day after infection, whereas uninfected BALB/c control animals had approximately 10- to 30-fold lower sag expression. Further fractionation experiments with small intestine indicated that sag expression occurred in gut-associated lymphoid cells. Restriction enzyme digestion of PCR products indicated that the sag mRNA detected was derived from the endogenous MMTVs, and sequencing analysis confirmed that the PCR products were derived from endogenous MTv-6. Expression of C3H-specific mRNA was detectable in BALB/cfC3H or C3H tissues by RNase protection or by RT-PCR. Endogenous MMTV sag expression was low in spleen and undetectable in thymocytes of C3H MMTV-infected C57BL/6 mice, a strain resistant to C3H MMTV tumorigenesis and defective for MHC class II I-E molecules. The RT-PCR assay for sag mRNA appears to measure the Sag-induced stimulation previously predicted for milk-borne MMTV infection. Together these data suggest that exogenous MMTV sag expression is minimal, but sufficient to rapidly stimulate transcription of endogenous MMTV sag mRNA in B- and T-cells in an MHC class II I-E-dependent manner. The endogenous sag expression on maternal lymphocytes may increase the number of proliferating T-cells available for milk-borne MMTV infection.


Subject(s)
Antigens, Viral/biosynthesis , Mammary Tumor Virus, Mouse/metabolism , Retroviridae Infections/virology , Superantigens/biosynthesis , Tumor Virus Infections/virology , Animals , Antigens, Viral/genetics , Base Sequence , Cloning, Molecular , DNA, Complementary , DNA, Viral , Female , Gene Expression Regulation, Viral , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Milk/virology , Molecular Sequence Data , RNA, Messenger/analysis , Retroviridae Infections/metabolism , Sequence Homology, Nucleic Acid , Superantigens/genetics , Tumor Virus Infections/metabolism
20.
Proc Natl Acad Sci U S A ; 92(11): 4828-32, 1995 May 23.
Article in English | MEDLINE | ID: mdl-7761408

ABSTRACT

Mouse mammary tumor virus (MMTV) encodes a superantigen that is important for viral infectivity in vivo. To determine whether superantigen function was required for infection by milk-borne MMTV, we created HYB PRO/Cla transgenic mice. These mice produced a full-length, packaged viral RNA with a frameshift mutation that caused premature termination of the superantigen protein. Young HYB PRO/Cla mice showed no deletion of their cognate V beta 14+ T cells, although they shed virus in their milk. The nontransgenic offspring of the HYB PRO/Cla mice were infected with this virus, since transgene-specific viral transcripts were detected in their mammary glands. Surprisingly, these offspring demonstrated the progressive deletion of V beta 14+ T cells characteristic of exogenous MMTV (C3H) infection. Sequence analysis demonstrated that these newly acquired viruses had reconstituted superantigen open reading frames resulting from recombination between the HYB PRO/Cla and endogenous Mtv-1 proviral RNAs. Thus, there is selection during the infection process for MMTVs with functional superantigen genes.


Subject(s)
Mammary Tumor Virus, Mouse/genetics , Repetitive Sequences, Nucleic Acid , Superantigens/genetics , Amino Acid Sequence , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Base Sequence , DNA Primers , Female , Flow Cytometry , Gene Library , Male , Mammary Tumor Virus, Mouse/immunology , Mammary Tumor Virus, Mouse/isolation & purification , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Mice, Transgenic , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , Recombination, Genetic , Superantigens/biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...