Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
2.
Oncogene ; 29(19): 2760-71, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20228844

ABSTRACT

Non-small cell lung cancers (NSCLC) that express the cell surface adhesion protein E-cadherin may carry a better prognosis than E-cadherin-negative tumors. Here, we found substantial inhibition of anchorage-independent growth in soft agar and cell migration in each of four NSCLC lines stably transfected with E-cadherin. The inhibitory effects were independent of the EGFR and beta-catenin/Wnt-signaling pathways. However, E-cadherin expression was associated with an adhesion-dependent reduction in the activity of Rho family proteins, RhoA in two lines and Cdc42 in the other two. The reduction of RhoA activity was dependent on DLC-1 Rho-GAP and p190 Rho-GAP and associated with an increase in a membrane-associated p190 Rho-GAP/p120 Ras-GAP complex. In parental cells with high levels of RhoA-GTP, siRNA-mediated knock-down of RhoA reduced cell migration and agar growth in a manner analogous to E-cadherin. In parental cells with high levels of Cdc42-GTP, transfection of a Cdc42 dominant-negative mutant reduced cell growth and migration similarly to cells expressing E-cadherin. Thus, E-cadherin can negatively regulate cell proliferation and migration in NSCLC by reducing the level of the predominant active form of Rho family protein, RhoA or Cdc42. These proteins can be considered downstream effectors of E-cadherin and might represent therapeutic targets in some NSCLC.


Subject(s)
Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , rho GTP-Binding Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
3.
Oncogene ; 28(11): 1401-9, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19151751

ABSTRACT

DLC1 (deleted in liver cancer 1), which encodes a Rho GTPase-activating protein (Rho-GAP), is a potent tumor suppressor gene that is frequently inactivated in several human cancers. DLC1 is a multidomain protein that has been shown previously to bind members of the tensin gene family. Here we show that p120Ras-GAP (Ras-GAP; also known as RASA1) interacts and extensively colocalizes with DLC1 in focal adhesions. The binding was mapped to the SH3 domain located in the N terminus of Ras-GAP and to the Rho-GAP catalytic domain located in the C terminus of the DLC1. In vitro analyses with purified proteins determined that the isolated Ras-GAP SH3 domain inhibits DLC1 Rho-GAP activity, suggesting that Ras-GAP is a negative regulator of DLC1 Rho-GAP activity. Consistent with this possibility, we found that ectopic overexpression of Ras-GAP in a Ras-GAP-insensitive tumor line impaired the growth-suppressing activity of DLC1 and increased RhoA activity in vivo. Our observations expand the complexity of proteins that regulate DLC1 function and define a novel mechanism of the cross talk between Ras and Rho GTPases.1R01CA129610


Subject(s)
Neoplasms/pathology , Tumor Suppressor Proteins/physiology , p120 GTPase Activating Protein/physiology , rhoA GTP-Binding Protein/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , GTPase-Activating Proteins , Humans , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , p120 GTPase Activating Protein/analysis , p120 GTPase Activating Protein/chemistry , src Homology Domains
5.
Expert Opin Biol Ther ; 1(4): 571-81, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11727495

ABSTRACT

Non-infectious human papillomavirus-like particles (VLP), composed of the L1 major capsid protein, are under active development as vaccines to prevent cervical cancer. They would presumably function primarily by generating virion-neutralising antibodies against the genital human papillomavirus (HPV) types that are the principal cause of most cervical cancers. Early phase clinical studies indicate that the VLP vaccines are well tolerated and able to consistently induce high titres of virus type-specific neutralising antibodies. Two types of second-generation VLP-based subunit vaccines with therapeutic implications, both related and unrelated to papillomavirus infection, are in preclinical development. One type seeks to induce cell-mediated immune responses, especially cytotoxic lymphocytes (CTL), against non-structural papillomavirus proteins, proteins of other viruses, or tumour associated antigens. The target antigen is incorporated into the VLPs as a fusion protein of L1 or the L2 minor capsid protein. In mouse models, this approach has generated potent CTL responses after low dose vaccination in the absence adjuvant. The second type of therapeutic VLP-based vaccine seeks to generate autoantibodies to self-antigens. The display of self polypeptides in the context of the highly ordered array of repetitive elements on the papillomavirus VLP surface abrogates the ability of the humoral immune system to functionally distinguish between foreign and self. High titre and high avidity auto-reactive IgG antibodies have been induced to both soluble (TNF-alpha) and cell surface (CCR5) central self-antigens. Vaccines based on this approach could potentially be effective alternatives to monoclonal antibody (mAb)-based therapies for a variety of disease targets.


Subject(s)
Capsid/immunology , Oncogene Proteins, Viral/immunology , Papillomaviridae/genetics , Papillomavirus Vaccines , Uterine Cervical Neoplasms/drug therapy , Viral Vaccines/therapeutic use , Adult , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , B-Lymphocytes/immunology , Capsid/genetics , Capsid Proteins , Clinical Trials as Topic , Dendritic Cells/immunology , Female , Humans , Oncogene Proteins, Viral/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control
6.
J Virol ; 75(21): 10493-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581419

ABSTRACT

Coexpression of bovine papillomavirus L1 with L2 mutants lacking either eight N-terminal or nine C-terminal amino acids that encode positively charged domains resulted in wild-type levels of viral genome encapsidation. Despite wild-type binding to the cell surface, the resulting virions were noninfectious. An L2 mutant encoding a scrambled version of the nine C-terminal residues restored infectivity, in contrast to an L2 mutant encoding a scrambled version of the N-terminal residues.


Subject(s)
Bovine papillomavirus 1/physiology , Capsid Proteins , Capsid/physiology , Nuclear Proteins , Animals , Capsid/chemistry , Cell Line , Cricetinae , Neoplasm Proteins/analysis , Transcription Factors/analysis , Tumor Suppressor Proteins , Virion/physiology , Virus Assembly
8.
J Clin Invest ; 108(3): 415-23, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489935

ABSTRACT

High avidity and long-lasting autoantibodies to a self-polypeptide (TNF-alpha) were generated after parenteral vaccination of mice with low doses of virus-like particle-based (VLP-based) vaccines that were constructed by linking mouse TNF-alpha peptides to the surface of papillomavirus VLPs. High-titer autoantibodies were induced with or without coadministration of potent conventional adjuvants, but were enhanced by coadministration of CFA. Compared with immunization with the fusion protein alone, attachment to VLPs increased autoantibody titers 1,000-fold. A comparison of Ab responses against the self (TNF-alpha) and foreign components of the fusion protein showed that VLP conjugation abrogated the ability of the humoral immune system to distinguish between self and foreign. Similar levels of IgM were detected to self and foreign epitopes regardless of the assembly state of the antigen, suggesting that conjugation of self-peptides to VLPs promotes survival or expansion of mature autoreactive B cells. In a mouse model, vaccination with conjugated particles inhibited development of type II collagen-induced arthritis. Together, these results suggest a potentially flexible method to efficiently generate autoantibodies against specific self-proteins that mediate arthritis and other diseases.


Subject(s)
Autoantibodies/biosynthesis , Autoantigens/administration & dosage , Papillomaviridae/immunology , Amino Acid Sequence , Animals , Arthritis/etiology , Arthritis/immunology , Arthritis/pathology , Arthritis/prevention & control , Autoantigens/genetics , Immunoglobulin M/biosynthesis , Male , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccination , Vaccines, Conjugate/administration & dosage
9.
J Immunol ; 166(9): 5346-55, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313370

ABSTRACT

The role of viral structural proteins in the initiation of adaptive immune responses is poorly understood. To address this issue, we focused on the effect of noninfectious papillomavirus-like particles (VLPs) on dendritic cell (DC) activation. We found that murine bone marrow-derived dendritic cells (BMDCs) effectively bound and rapidly internalized bovine papillomavirus VLPS: Exposure to fully assembled VLPs of bovine papillomavirus, human papillomavirus (HPV)16 or HPV18, but not to predominately disordered HPV16 capsomers, induced acute phenotypic maturation of BMDCS: Structurally similar polyomavirus VLPs bound to the DC surface and were internalized, but failed to induce maturation. DCs that had incorporated HPV16 VLPs produced proinflammatory cytokines IL-6 and TNF-alpha; however, the release of these cytokines was delayed relative to LPS activation. Production of IL-12p70 by VLP-exposed DCs required the addition of syngeneic T cells or rIFN-gamma. Finally, BMDCs pulsed with HPV16 VLPs induced Th1-dominated primary T cell responses in vitro. Our data provide evidence that DCs respond to intact papillomavirus capsids and that they play a central role in VLP-induced immunity. These results offer a mechanistic explanation for the striking ability of papillomavirus VLP-based vaccines to induce potent T and B cell responses even in the absence of adjuvant.


Subject(s)
Bovine papillomavirus 1/immunology , Capsid Proteins , Dendritic Cells/immunology , Dendritic Cells/virology , Papillomaviridae/immunology , Virion/immunology , Animals , BK Virus/immunology , Capsid/immunology , Capsid/metabolism , Cattle , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Interphase/immunology , JC Virus/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Protein Binding/immunology , Th1 Cells/immunology , Th1 Cells/virology , Virus Assembly/genetics , Virus Assembly/immunology
10.
J Virol ; 75(9): 4332-42, 2001 May.
Article in English | MEDLINE | ID: mdl-11287582

ABSTRACT

BPHE-1 cells, which harbor 50 to 200 viral episomes, encapsidate viral genome and generate infectious bovine papillomavirus type 1 (BPV1) upon coexpression of capsid proteins L1 and L2 of BPV1, but not coexpression of BPV1 L1 and human papillomavirus type 16 (HPV16) L2. BPV1 L2 bound in vitro via its C-terminal 85 residues to purified L1 capsomers, but not with intact L1 virus-like particles in vitro. However, when the efficiency of BPV1 L1 coimmunoprecipitation with a series of BPV1 L2 deletion mutants was examined in vivo, the results suggested that residues 129 to 246 and 384 to 460 contain independent L1 interaction domains. An L2 mutant lacking the C-terminal L1 interaction domain was impaired for encapsidation of the viral genome. Coexpression of BPV1 L1 and a chimeric L2 protein composed of HPV16 L2 residues 1 to 98 fused to BPV1 L2 residues 99 to 469 generated infectious virions. However, inefficient encapsidation was seen when L1 was coexpressed with either BPV1 L2 with residues 91 to 246 deleted or with BPV1 L2 with residues 1 to 225 replaced with HPV16 L2. Impaired genome encapsidation did not correlate closely with impairment of the L2 proteins either to localize to promyelocytic leukemia oncogenic domains (PODs) or to induce localization of L1 or E2 to PODs. We conclude that the L1-binding domain located near the C terminus of L2 may bind L1 prior to completion of capsid assembly, and that both L1-binding domains of L2 are required for efficient encapsidation of the viral genome.


Subject(s)
Bovine papillomavirus 1/metabolism , Capsid Proteins , Capsid/metabolism , Genome, Viral , Virus Assembly/physiology , Animals , Binding Sites , Bovine papillomavirus 1/genetics , Capsid/genetics , Cattle , Cell Line , Cricetinae , Humans , Mutagenesis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomaviridae/metabolism , Precipitin Tests
11.
Cancer Epidemiol Biomarkers Prev ; 10(2): 101-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11219765

ABSTRACT

Genital human papillomavirus (HPV) infection is generally considered to be sexually transmitted. However, nonsexual spread of the virus has also been suggested. The goal of this study was to assess: (a) the role of sexual intercourse in the transmission of HPV; (b) the determinants for seroconversion; and (c) the correlation between HPV DNA, abnormal cervical cytology, and serological response to HPV16. One hundred virgins and 105 monogamous women were randomly selected from a population-based cohort study in Copenhagen, Denmark, in which the women were examined twice with 2-year interval (interview, cervical swabs, Pap smear, blood samples). The presence of HPV DNA was determined by GP5+/6+ primers based HPV-PCR-EIA. HPV 16 virus-like particles (VLP) antibodies were detected by ELISA. All of the virgins were both HPV DNA negative and seronegative to VLP16, except for one woman who was weakly HPV 6 DNA positive. Only those virgins who initiated sexual activity became HPV DNA positive and/or VLP16 positive. The most important determinant of HPV DNA acquisition was the number of partners between the two examinations. The only significant risk factor for HPV 16 VLP seroconversion among women acquiring HPV DNA was HPV type. Our results show that sexual intercourse is important in the transmission of HPV, and that HPV 16 VLP seroconversion and the development of cervical lesions only occur after HPV transmission. Remarkably, no cervical lesions were found in HPV 16 DNA positive women who had seroconverted. Although based on small numbers, this may suggest that the development of antibodies had a protective effect.


Subject(s)
Coitus , Disease Transmission, Infectious/statistics & numerical data , Papillomaviridae/isolation & purification , Papillomavirus Infections/transmission , Sexually Transmitted Diseases, Viral/transmission , Tumor Virus Infections/transmission , Adult , Cohort Studies , DNA, Viral/analysis , Denmark/epidemiology , Female , Humans , Incidence , Longitudinal Studies , Papillomavirus Infections/diagnosis , Papillomavirus Infections/epidemiology , Polymerase Chain Reaction/methods , Prospective Studies , Risk Assessment , Risk Factors , Sexually Transmitted Diseases, Viral/diagnosis , Sexually Transmitted Diseases, Viral/epidemiology , Tumor Virus Infections/diagnosis , Tumor Virus Infections/epidemiology
12.
J Natl Cancer Inst ; 93(4): 284-92, 2001 Feb 21.
Article in English | MEDLINE | ID: mdl-11181775

ABSTRACT

BACKGROUND: Studies in animal models have shown that systemic immunization with a papillomavirus virus-like particle (VLP) vaccine composed of L1, a major structural viral protein, can confer protection against subsequent experimental challenge with the homologous virus. Here we report results of a double-blind, placebo-controlled, dose-escalation trial to evaluate the safety and immunogenicity of a human papillomavirus (HPV) type 16 (HPV16) L1 VLP vaccine in healthy adults. METHODS: Volunteers were given intramuscular injections with placebo or with 10- or 50-microg doses of HPV16 L1 VLP vaccine given without adjuvant or with alum or MF59 as adjuvants at 0, 1, and 4 months. All vaccine recipients were monitored for clinical signs and symptoms for 7 days after each inoculation. Immune responses were measured by an HPV16 L1 VLP-based enzyme-linked immunosorbent assay (ELISA) and by an HPV16 pseudovirion neutralization assay. The antibody titers were given as the reciprocals of the highest dilution showing positive reactivity in each assay. All statistical tests were two-sided. RESULTS: The prevaccination geometric mean ELISA titer for six seropositive individuals was 202 (range, 40--640). All vaccine formulations were well tolerated, and all subjects receiving vaccine seroconverted. Serum antibody responses at 1 month after the third injection were dose dependent in recipients of vaccine without adjuvant or with MF59 but were similar at both doses when alum was the adjuvant. With the higher dose, the geometric means of serum ELISA antibody titers (95% confidence intervals) to purified VLP 1 month after the third injection were as follows: 10,240 (1499 to 69 938) without adjuvant, 10,240 (1114 to 94 145) with MF59, and 2190 (838 to 5723) with alum. Responses of subjects within each group were similar. Neutralizing and ELISA antibody titers were highly correlated (Spearman correlation =.85), confirming that ELISA titers are valid proxies for neutralizing antibodies. CONCLUSIONS: The HPV16 L1 VLP vaccine is well tolerated and is highly immunogenic even without adjuvant, with the majority of the recipients achieving serum antibody titers that were approximately 40-fold higher than what is observed in natural infection.


Subject(s)
Antibodies, Viral/blood , Papillomaviridae/immunology , Papillomavirus Vaccines , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Alum Compounds/administration & dosage , Baculoviridae , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization Schedule , Immunoglobulins/blood , Injections, Intramuscular , Male , Polysorbates/administration & dosage , Recombinant Proteins , Reference Values , Squalene/administration & dosage , Viral Vaccines/administration & dosage
13.
J Natl Cancer Inst Monogr ; (28): 50-4, 2001.
Article in English | MEDLINE | ID: mdl-11158207

ABSTRACT

Papillomavirus-like particle (VLP)-based subunit vaccines have undergone rapid development over the past 8 years. Three types are being investigated. The most basic type is composed of only the L1 major capsid protein and is designed to prevent genital human papillomavirus (HPV) infection by inducing virus-neutralizing antibodies. On the basis of positive results in animal models, clinical trials of this type of vaccine for HPV16, and other types, are currently under way. Preliminary results have been encouraging in that systemic immunization with the L1 VLPs induced high serum titers of neutralizing antibodies without substantial adverse effects. The second type of vaccine incorporates other papillomavirus polypeptides into the VLPs as L1 or L2 fusion proteins. These chimeric VLPs are designed to increase the therapeutic potential of an HPV vaccine by inducing cell-mediated responses to nonstructural viral proteins, such as E7. Studies in mice indicate that these vaccines generate potent antitumor cytotoxic lymphocyte (CTL) responses while retaining the ability to induce high-titer neutralizing antibodies. It is likely that prophylactic and therapeutic clinical trials of chimeric VLPs will be initiated in the near future. The third type of VLP-based vaccine is designed to induce autoantibodies against central self-antigens by incorporating self-peptides into the outer surface of VLPs, a process that could have therapeutic potential in various disease settings unrelated to HPV infection. In a recent proof of concept study, a peptide from an external loop of mouse CCR5 protein was inserted into a neutralizing epitope of L1. In mice, the particles generated by this chimeric L1 were able to induce high titers of CCR5 antibodies that specifically recognized the surface of CCR5-transfected cells and blocked in vitro infection of an M-tropic human immunodeficiency virus strain.


Subject(s)
Capsid Proteins , Papillomaviridae/immunology , Papillomavirus Vaccines , Vaccines, Subunit , Viral Vaccines , Animals , Antibodies, Viral/biosynthesis , Autoantibodies/biosynthesis , Clinical Trials as Topic , Cytotoxicity, Immunologic , Female , HIV Infections/prevention & control , Humans , Immunization , Male , Mice , Oncogene Proteins, Viral/immunology , Papillomaviridae/ultrastructure , Papillomavirus Infections/prevention & control , Rabbits , Receptors, CCR5/immunology , Receptors, HIV/immunology , Recombinant Fusion Proteins/immunology , Sexually Transmitted Diseases, Viral/prevention & control , Transfection , Tumor Virus Infections/prevention & control , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Virion/ultrastructure
14.
Virology ; 279(1): 361-9, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145917

ABSTRACT

Genital HPV genotypes are generally distinct serotypes, but whether variants within a genotype can represent serologic subtypes is unclear. In this study we used serum from human volunteers vaccinated with HPV16 L1 VLPs from variant 114K, to examine cross-neutralization of variants from each of the five major phylogenetic branches of HPV16. Recombinant Semliki Forest virus-derived pseudovirions for each variant were generated and combined with serum from vaccines, and the mixture was monitored for infectivity in a standard C127 cell focal transformation assay. Sera from all 10 VLP-immunized individuals had neutralizing activity against each of the variant pseudovirions. For each of the sera, variant titers differed by only fourfold or less from the median titer. Therefore, from a vaccine perspective, HPV16 variants belong to a single serotype. Vaccination with HPV16 114K L1 VLPs generates antibodies that should confer a similar degree of protection against all known phylogenetic branches of HPV16.


Subject(s)
Antibodies, Viral/immunology , Papillomaviridae/immunology , Viral Vaccines/immunology , Virion/immunology , Antibodies, Viral/blood , Cross Reactions , Humans , Immunization , Neutralization Tests , Papillomaviridae/classification , Papillomaviridae/genetics , Papillomavirus Infections/prevention & control , Phylogeny , Serotyping , Tumor Virus Infections/prevention & control , Viral Vaccines/administration & dosage , Virion/metabolism , Virus Assembly
15.
Virology ; 270(2): 254-7, 2000 May 10.
Article in English | MEDLINE | ID: mdl-10792983

ABSTRACT

Vaccination with virus-like particles (VLP), comprising both L1 and L2 of human papillomavirus (HPV) genital types 6, 16, and 18, induces predominantly type-specific neutralizing antibodies. L2 polypeptide vaccines protect animals against experimental challenge with homologous papillomavirus and cross-reactive epitopes are present in HPV L2. To assess L2-specific cross-neutralization of HPV genotypes, sheep were immunized with purified, bacterially expressed HPV6, 16, or 18 L2. In addition to neutralizing the homologous HPV type in vitro, antisera to each HPV L2 also cross-neutralized both heterologous HPV types. This suggests that unlike VLP-based prophylactic HPV vaccines, an L2 polypeptide vaccine may provide broad-spectrum protection.


Subject(s)
Capsid/immunology , Immunodominant Epitopes/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Tumor Virus Infections/immunology , Viral Vaccines/immunology , Cross Reactions , Epitopes/immunology , Humans , Papillomavirus Infections/prevention & control , Tumor Virus Infections/prevention & control
16.
EMBO J ; 19(4): 642-54, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675333

ABSTRACT

Targeted disruption of both alleles of mouse sos1, which encodes a Ras-specific exchange factor, conferred mid-gestational embryonic lethality that was secondary to impaired placental development and was associated with very low placental ERK activity. The trophoblastic layers of sos1(-/-) embryos were poorly developed, correlating with high sos1 expression in wild-type trophoblasts. A sos1(-/-) cell line, which expressed readily detectable levels of the closely related Sos2 protein, formed complexes between Sos2, epidermal growth factor receptor (EGFR) and Shc efficiently, gave normal Ras.GTP and ERK responses when treated with EGF for < or =10 min and was transformed readily by activated Ras. However, the sos1(-/-) cells were resistant to transformation by v-Src or by overexpressed EGFR and continuous EGF treatment, unlike sos1(+/-) or wild-type cells. This correlated with Sos2 binding less efficiently than Sos1 to EGFR and Shc in cells treated with EGF for > or =90 min or to v-Src and Shc in v-Src-expressing cells, and with less ERK activity. We conclude that Sos1 participates in both short- and long-term signaling, while Sos2-dependent signals are predominantly short-term.


Subject(s)
Placenta/metabolism , SOS1 Protein/genetics , SOS1 Protein/metabolism , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , ras Proteins/metabolism , Animals , Base Sequence , Cell Line, Transformed , DNA Primers/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression , Genes, ras , Genes, src , In Situ Hybridization , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Pregnancy , Signal Transduction , ras Proteins/genetics
17.
Mol Cell Biol ; 19(7): 4611-22, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373510

ABSTRACT

The full-length versions of the Ras-specific exchange factors Ras-GRF1 (GRF1) and Ras-GRF2 (GRF2), which are expressed in brain and a restricted number of other organs, possess an ionomycin-dependent activation of Erk mitogen-activated protein kinase activity in 293T cells (C. L. Farnsworth et al., Nature 376:524-527, 1995; N. P. Fam et al., Mol. Cell. Biol. 17:1396-1406, 1996). Each GRF protein contains a Dbl homology (DH) domain. A yeast two-hybrid screen was used to identify polypeptides that associate with the DH domain of GRF1. In this screen, a positive cDNA clone from a human brain cDNA library was isolated which consisted of the GRF2 DH domain and its adjacent ilimaquinone domain. Deletion analysis verified that the two-hybrid interaction required only the DH domains, and mutation of Leu-263 to Gln (L263Q) in the N terminus of the GRF1 DH domain abolished the two-hybrid interaction, while a cluster of more C-terminally located mutations in the DH domain did not eliminate the interaction. Oligomers between GRF1 and GRF2 were detected in a rat brain extract, and forced expression of GRF1 and GRF2 in cultured mammalian cells formed homo- and hetero-oligomers. Introduction of the L263Q mutation in GRF1 led to a protein that was deficient in oligomer formation, while GRF1 containing the DH cluster mutations formed homo-oligomers with an efficiency similar to that of wild type. Compared to wild-type GRF1, the focus-forming activity on NIH 3T3 cells of the GRF1 DH cluster mutant was reduced, while the L263Q mutant was inactive. Both mutants were impaired in their ability to mediate ionomycin-dependent Erk activity in 293T cells. In the absence of ionomycin, 293T cells expressing wild-type GRF1 contained much higher levels of Ras-GTP than control cells; the increase in Erk activity induced by ionomycin in the GRF1-expressing cells also induced a concomitant increase in Raf kinase activity, but without a further increase in the level Ras-GTP. We conclude that GRF1 and GRF2 can form homo- and hetero-oligomers via their DH domains, that mutational inactivation of oligomer formation by GRF1 is associated with impaired biological and signaling activities, and that in 293T cells GRF1 mediates at least two pathways for Raf activation: one a constitutive signal that is mainly Ras-dependent, and one an ionomycin-induced signal that cooperates with the constitutive signal without further augmenting the level of GTP-Ras.


Subject(s)
Calcium/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Transformed , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Enzyme Activation , Guanine Nucleotide Exchange Factors , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Mice , Molecular Sequence Data , Oligopeptides/genetics , Oligopeptides/metabolism , Proteins/genetics , Rats , Sequence Homology, Amino Acid , ras Guanine Nucleotide Exchange Factors , ras-GRF1
18.
Proc Natl Acad Sci U S A ; 96(5): 2373-8, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051649

ABSTRACT

The vertebrate immune system has evolved to respond vigorously to microbial infection but to ignore self-antigens. Evidence has emerged that B cell responses to viruses are initiated by immune recognition of ordered arrays of antigen on the viral surface. To test whether autoantibodies against a self-antigen can be induced by placing it in a context that mimics the ordered surface of a viral particle, a peptide representing an extracellular loop of the mouse chemokine receptor CCR5 was incorporated into an immunodominant site of the bovine papillomavirus virus L1 coat protein, which self-assembles into virus-like particles. Mice inoculated with chimeric L1-CCR5 particles generated autoantibodies that bound to native mouse CCR5, inhibited binding of its ligand RANTES, and blocked HIV-1 infection of an indicator cell line expressing a human-mouse CCR5 chimera. These results suggest a general method for inducing autoantibodies against self-antigens, with diverse potential basic research and clinical applications.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Immunoglobulin G/blood , Receptors, CCR5/immunology , Animals , Antibody Formation , Bovine papillomavirus 1/genetics , Capsid/genetics , Capsid/immunology , Cattle , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, CCR5/chemistry , Recombinant Fusion Proteins/immunology
19.
Biochim Biophys Acta ; 1423(1): M1-8, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-9989208

ABSTRACT

Identification of a subset of HPV types as etiologic agents of cervical cancer and other malignancies implies that development of an effective vaccine against HPV infection could have a major impact on tumors attributable to these viruses. The ability of the L1 major capsid protein of papillomaviruses to self-assemble into VLPs that can, when inoculated systemically, induce high levels of neutralizing antibodies and protect animals against experimental viral challenge makes L1 VLPs an excellent candidate subunit vaccine. VLPs have the limitation of inducing type-specific immunity. Studies in humans are required to determine whether systemic vaccination with L1 VLPs will prevent sexually transmitted HPV infection. Since prospective efficacy trials will take several years to complete, considering alternative approaches is also worthwhile.


Subject(s)
Papillomaviridae , Papillomavirus Vaccines , Tumor Virus Infections/prevention & control , Uterine Cervical Neoplasms/prevention & control , Viral Vaccines/administration & dosage , Animals , Capsid/chemistry , Capsid/immunology , Capsid Proteins , Chimera , Female , Humans , Oncogene Proteins, Viral/immunology , Risk Factors , Tumor Virus Infections/virology , Uterine Cervical Neoplasms/virology , Viral Vaccines/immunology , Virion/genetics , Virion/immunology
20.
Virology ; 250(2): 371-6, 1998 Oct 25.
Article in English | MEDLINE | ID: mdl-9792847

ABSTRACT

A single injection of HPV16 L1 virus-like particles induced potent CD8-mediated protection from tumor challenge by C3 cells, a line derived from embryonic mouse cells transfected with the HPV16 genome. L1 RNA, but not protein, was detected biochemically in C3 cells. These results indicate that low-level expression of HPV16 L1 can occur in proliferating cells and serve as a tumor vaccine target. Although L1 expression is generally thought to be restricted to terminally differentiated epithelial cells, these results suggest that additional analysis for low-level L1 expression in proliferating cells of HPV-induced lesions is warranted and might help in predicting the clinical potential of HPV L1 virus-like particle-based vaccines.


Subject(s)
Capsid Proteins , Oncogene Proteins, Viral/immunology , Papillomaviridae/immunology , Papillomavirus Infections/prevention & control , Tumor Virus Infections/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid/genetics , Capsid/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Tumor Cells, Cultured , Virion/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...