Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Biochem Biophys Res Commun ; 524(4): 825-831, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32037086

ABSTRACT

Chromatin organization starts from a "beads-on-a string" 10 nm fiber, a basic nucleosomal structure consisting of DNA and core histones. Given its regular nucleosome array on DNA backbone where N-terminal tails of each histone are exposed on the surface of chromatin fiber, we hypothesized that chromatin can be utilized as a heterologous peptide carrier to elicit a peptide-specific immune response. The plasmid DNA containing the Widom's clone 601 sequence and the recombinant chimeric histones containing the peptide derived from ras oncogene (G12V) were used to assemble the chromatin fiber in vitro. The immunogenicity of the assembled chromatin was tested in mice as a single vaccine component or formulated with adjuvants. G12V tagged-chromatin co-administered with adjuvants induced higher antibody responses against the G12V peptide than vaccination with adjuvant alone, while chimeric histones did not generate a significant antibody response. Interestingly, splenocytes from mice vaccinated with the G12V tagged-chromatin vaccine did not generate significant antigen-specific cytokine responses. Our studies suggest that chromatin can be utilized as an effective carrier of antigenic peptides for inducing specific antibody responses.


Subject(s)
Cancer Vaccines/biosynthesis , Genes, ras/immunology , Histones/immunology , Nanofibers/chemistry , Peptide Library , Peptides/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies/genetics , Antibodies/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Chromatin Assembly and Disassembly , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/prevention & control , Nucleosomes/chemistry , Nucleosomes/immunology , Nucleosomes/metabolism , Peptides/genetics , Peptides/metabolism , Plasmids/chemistry , Plasmids/immunology , Plasmids/metabolism , Vaccines, Subunit , Xenopus laevis
2.
Fish Shellfish Immunol ; 89: 281-289, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30953781

ABSTRACT

H-Ras is a guanosine triphosphatase (GTPase), which acts as a molecular switch and controls multiple important cellular processes including lymphocyte activation and function. However, regulatory mechanism of adaptive immune response by H-Ras remains unclear in non-mammalian animals. In the present study, we investigated the involvement of H-Ras in lymphocyte activation with a teleost model Oreochromis niloticus. H-Ras from O. niloticus (On-H-Ras) is highly conserved with those from other vertebrates. The mRNA of On-H-Ras showed a wide expression pattern in the lymphoid-tissues and with the highest level in liver. After Aeromonas hydrophila infection, transcription of On-H-Ras was significantly induced on day 8 but came back to basal level on day 16, suggesting that On-H-Ras potentially participated in primary response during the adaptive immunity. Furthermore, On-H-Ras mRNA was obviously up-regulated when leukocytes were activated by T lymphocyte mitogen PHA in vitro. Meanwhile, protein level of H-Ras was also augmented once leukocytes were stimulated with lymphocyte receptor signaling agonist PMA and ionomycin. More importantly, once Ras activity was inhibited by specific inhibitor, the up-regulation of lymphocyte activation marker CD122 was obviously impaired during lymphocyte activation process. Therefore, On-H-Ras regulated lymphocyte activation through both mRNA and protein level. Altogether, our results illustrated the involvement of H-Ras in teleost adaptive immunity via controlling lymphocyte activation, and thus provided a novel perspective to understand evolution of the lymphocyte-mediated adaptive immunity.


Subject(s)
Adaptive Immunity/genetics , Cichlids/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Genes, ras/immunology , Lymphocyte Activation/genetics , Aeromonas hydrophila/physiology , Animals , Cichlids/immunology , Fish Proteins/metabolism , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary
3.
J Clin Immunol ; 35(5): 454-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896945

ABSTRACT

RAS-associated leukoproliferative disease (RALD) is a newly classified disease; thus its clinical features and management are not fully understood. The cases of two patients with characteristic features of RALD are described herein. Patient 1 was a 5-month-old female with clinical features typical of autoimmune lymphoproliferative syndrome (ALPS) and markedly elevated TCRαß(+)CD4(-)CD8(-) T cell numbers. Genetic analyses failed to detect an ALPS-related gene mutation; however, whole exome sequencing and other genetic analyses revealed somatic mosaicism for the G13D NRAS mutation. These data were indivative of NRAS-associated RALD with highly elevated αß-double-negative T cells. Patient 2 was a 12-month-old girl with recurrent fever who clearly met the diagnostic criteria for juvenile myelomonocytic leukemia (JMML). Genetic analyses revealed somatic mosaicism, again for the G13D NRAS mutation, suggesting RALD associated with somatic NRAS mosaicism. Notably, unlike most JMML cases, Patient 2 did not require steroids or hematopoietic stem cell transplantation. Genetic analysis of RAS should be performed in patients fulfilling the diagnostic criteria for ALPS in the absence of ALPS-related gene mutations if the patients have elevated αß-double-negative-T cells and in JMML patients if autoimmunity is detected. These clinical and experimental data increase our understanding of RALD, ALPS, and JMML.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/immunology , GTP Phosphohydrolases/genetics , Genes, ras/immunology , Membrane Proteins/genetics , Mosaicism , T-Lymphocytes/physiology , Autoimmune Lymphoproliferative Syndrome/genetics , Female , Genetic Testing , Humans , Infant , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/immunology , Mutation/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism
4.
J Immunol ; 194(6): 2683-95, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25694611

ABSTRACT

Prevention of tumors induced by environmental carcinogens has not been achieved. Skin tumors produced by polyaromatic hydrocarbons, such as 7,12-dimethylbenz(a)anthracene (DMBA), often harbor an H-ras point mutation, suggesting that it is a poor target for early immunosurveillance. The application of pyrosequencing and allele-specific PCR techniques established that mutations in the genome and expression of the Mut H-ras gene could be detected as early as 1 d after DMBA application. Further, DMBA sensitization raised Mut H-ras epitope-specific CTLs capable of eliminating Mut H-ras(+) preneoplastic skin cells, demonstrating that immunosurveillance is normally induced but may be ineffective owing to insufficient effector pool size and/or immunosuppression. To test whether selective pre-expansion of CD8 T cells with specificity for the single Mut H-ras epitope was sufficient for tumor prevention, MHC class I epitope-focused lentivector-infected dendritic cell- and DNA-based vaccines were designed to bias toward CTL rather than regulatory T cell induction. Mut H-ras, but not wild-type H-ras, epitope-focused vaccination generated specific CTLs and inhibited DMBA-induced tumor initiation, growth, and progression in preventative and therapeutic settings. Transferred Mut H-ras-specific effectors induced rapid tumor regression, overcoming established tumor suppression in tumor-bearing mice. These studies support further evaluation of oncogenic mutations for their potential to act as early tumor-specific, immunogenic epitopes in expanding relevant immunosurveillance effectors to block tumor formation, rather than treating established tumors.


Subject(s)
Cancer Vaccines/therapeutic use , Genes, ras/genetics , Point Mutation/genetics , Skin Neoplasms/prevention & control , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Carcinogens/toxicity , Cytokines/immunology , Cytokines/metabolism , DNA Mutational Analysis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes/genetics , Epitopes/immunology , Female , Genes, ras/immunology , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Mice, Inbred C3H , Mice, Inbred Strains , Point Mutation/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Treatment Outcome , Tumor Burden/immunology
5.
Curr Opin Pediatr ; 25(6): 722-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24240292

ABSTRACT

PURPOSE OF REVIEW: Several autoimmune lymphoproliferative syndromes have been described lately. We review here the main clinical and laboratory findings of these new disorders. RECENT FINDINGS: The prototypical autoimmune lymphoproliferative syndrome (ALPS) has had its diagnostic criteria modified, somatic mutations in RAS genes were found to cause an ALPS-like syndrome in humans, and mutations in a gene encoding a protein kinase C (PRKCD) were discovered to cause a syndrome of lymphoproliferation, autoimmunity and natural killer cell defect. SUMMARY: The recent discoveries shed light on the molecular pathways governing lymphocyte death, proliferation and immune tolerance in humans.


Subject(s)
Apoptosis/immunology , Autoimmune Lymphoproliferative Syndrome/diagnosis , Genes, ras , Immune Tolerance/immunology , Inflammation/immunology , Apoptosis/genetics , Autoimmune Lymphoproliferative Syndrome/genetics , Autoimmune Lymphoproliferative Syndrome/immunology , Autoimmunity/genetics , Cell Proliferation , Fas-Associated Death Domain Protein/immunology , Female , Genes, ras/genetics , Genes, ras/immunology , Humans , Immune Tolerance/genetics , Inflammation/genetics , Inflammation/pathology , Male , Mutation/immunology , Protein Kinase C-delta/immunology , Signal Transduction/genetics , Signal Transduction/immunology
6.
Blood ; 117(19): 5102-11, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21444916

ABSTRACT

The small guanine nucleotide binding proteins of the Ras family, including in mammals the highly homologous H-ras, N-ras, and K-ras isoforms, are rapidly activated on ligation of the T-cell antigen receptor (TCR), but whether each isoform plays specific roles in T cells is largely unknown. Here, we show, with the use of mice specifically lacking H-ras or N-ras, that these isoforms are dispensable for thymocyte development and mature T-cell activation. By contrast, CD4⁺ T cells from Ras-deficient mice exhibited markedly decreased production of the Th1 signature cytokine IFN-γ early after TCR stimulation, concomitantly with impaired induction of the Th1-specific transcription factor T-bet. Accordingly, Ras-deficient mice failed to mount a protective Th1 response in vivo against the intracellular parasite Leishmania major, although they could be rendered resistant to infection if a Th1-biased milieu was provided during parasite challenge. Collectively, our data indicate that the TCR recruits distinct Ras isoforms for signal transduction in developing and mature T cells, thus providing a mechanism for differential signaling from the same surface receptor. Furthermore, we demonstrate for the first time that H-ras and N-ras act as critical controllers of Th1 responses, mostly by transmitting TCR signals for Th1 priming of CD4⁺ T cells.


Subject(s)
Cell Differentiation/genetics , Genes, ras/immunology , Lymphocyte Activation/genetics , Signal Transduction/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Immunoblotting , Leishmaniasis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Th1 Cells/cytology
7.
Int J Cancer ; 128(5): 1120-8, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-20473937

ABSTRACT

K-ras mutations are frequently found in adenocarcinomas of the pancreas and can elicit mutation-specific immune responses. Targeting the immune system against mutant Ras may thus influence the clinical course of the disease. Twenty-three patients who were vaccinated after surgical resection for pancreatic adenocarcinoma (22 pancreaticoduodenectomies, one distal resection), in two previous Phase I/II clinical trials, were followed for more than 10 years with respect to long-term immunological T-cell reactivity and survival. The vaccine was composed of long synthetic mutant ras peptides designed mainly to elicit T-helper responses. Seventeen of 20 evaluable patients (85%) responded immunologically to the vaccine. Median survival for all patients was 27.5 months and 28 months for immune responders. The 5-year survival was 22% and 29%, respectively. Strikingly, 10-year survival was 20% (four patients out of 20 evaluable) versus zero (0/87) in a cohort of nonvaccinated patient treated in the same period. Three patients mounted a memory response up to 9 years after vaccination. The present observation of long-term immune response together with 10-year survival following surgical resection indicates that K-ras vaccination may consolidate the effect of surgery and represent an adjuvant treatment option for the future.


Subject(s)
Adenocarcinoma/immunology , Cancer Vaccines/administration & dosage , Genes, ras/immunology , Pancreatic Neoplasms/immunology , Adenocarcinoma/surgery , Adenocarcinoma/therapy , Aged , Female , Follow-Up Studies , Humans , Hypersensitivity, Delayed , Immunologic Memory , Male , Middle Aged , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/therapy , Survival Analysis , T-Lymphocytes/immunology
8.
Am J Clin Oncol ; 34(3): 321-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20686403

ABSTRACT

PURPOSE: Codon 12 mutations of K-ras are frequent in pancreatic adenocarcinoma, and represent potential tumor-specific neoantigens. The objective of this study was to assess the safety and efficacy of immunizing patients with resected pancreatic cancer with a vaccine targeted against their tumor-specific K-ras mutation. METHODS: Patients with resected pancreatic cancer, with K-ras mutations at codon 12, were vaccinated once monthly for 3 months with a 21-mer peptide vaccine containing the corresponding K-ras mutation of the patient's tumor. About 200 µg of peptide vaccine was injected intradermally on day 7 of a 10-day course of intradermal granulocyte macrophage colony-stimulating factor. Toxicity was assessed by National Cancer Institute Common Toxicity Criteria v2.0. Immune responses were evaluated by delayed-type hypersensitivity (DTH) tests and the enzyme-linked immunosorbent spot assays. RESULTS: Of 62 screened patients, 24 were vaccinated. There were no grade 3-5 vaccine-specific toxicities. The only National Cancer Institute grade 1 and 2 toxicity was erythema at the injection site (94%). Nine patients (25%) were evaluable for immunologic responses. One patient (11%) had a detectable immune response specific to the patient's K-ras mutation, as assessed by DTH. Three patients (13%) displayed a DTH response that was not specific. Median recurrence free survival time was 8.6 months (95% confidence interval, 2.96-19.2) and median overall survival time was 20.3 months (95% confidence interval, 11.6-45.3). CONCLUSIONS: K-ras vaccination for patients with resectable pancreatic adenocarcinoma proved to be safe and tolerable with however no elicitable immunogenicity and unproven efficacy. Future development of adjuvant vaccine therapies should use more immunogenic vaccines.


Subject(s)
Adenocarcinoma/drug therapy , Cancer Vaccines/therapeutic use , Genes, ras/immunology , Mutation/immunology , Pancreatectomy , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/surgery , Adult , Aged , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Confounding Factors, Epidemiologic , Drug Administration Schedule , Enzyme-Linked Immunospot Assay , Feasibility Studies , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Hypersensitivity, Delayed/diagnosis , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/surgery , Time Factors , Treatment Outcome
9.
J Immunol Methods ; 277(1-2): 27-37, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12799037

ABSTRACT

The induction and repair of DNA damage has been shown to occur heterogeneously throughout the mammalian genome. As a consequence, analysis of these parameters at a global genome level may not reflect important gene-level events. Few techniques have been established to explore quantitatively gene-specific DNA damage and repair. Most of these are polymerase chain reaction (PCR)-based assays and are relatively insensitive, relying on decreased PCR amplification arising from damage in template DNA. We have developed a quantitative assay that combines specific immunocapture of damaged DNA by an antiserum specific for thymine dimers (IgG479), with PCR amplification of a 149 bp fragment of the human H-ras proto-oncogene. Quantification of DNA damage was based upon proportionality between the amount of the PCR product and the initial amount of damage. Detection of thymine dimers was possible with nanogram amounts of genomic DNA and increased in a linear, dose-responsive manner. Using this assay, gene-level induction of thymine dimers was shown to be directly proportional to levels induced in the global genome of ultraviolet radiation (UVR)-exposed, extracted DNA as measured by gas chromatography-mass spectrometry (GC-MS). This result suggests that global damage assessments do indeed reflect gene-level events although we predict that this relationship may not be maintained when applied to a cellular system. These findings demonstrate the suitability of this approach to the detection of UVR-induced DNA damage at the level of individual genes.


Subject(s)
DNA Damage , DNA Damage/immunology , Immunoglobulin G/immunology , Polymerase Chain Reaction/methods , Pyrimidine Dimers/analysis , Ultraviolet Rays/adverse effects , Antibody Specificity , DNA Damage/genetics , Enzyme-Linked Immunosorbent Assay , Genes, ras/genetics , Genes, ras/immunology , Genes, ras/radiation effects , Humans , Poly T/immunology , Poly T/radiation effects , Proto-Oncogene Mas , Pyrimidine Dimers/genetics
10.
Cancer Res ; 63(7): 1615-22, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12670913

ABSTRACT

Ras proteins have a key role in the regulation of several cellular functions, and are involved in a significant percentage of human tumors. However, the specific functions of the different Ras isoforms are poorly understood. In this work, we show for the first time a specific role for N-ras in T-cell function and development. Mice defective for N-ras have low numbers of CD8 single positive thymocytes and decreased thymocyte proliferation in vitro. In Ras signaling and activation assays, KO-N-ras thymocytes showed a defective response to T-cell activation. In turn, these deficiencies resulted in a significant reduction in the production of interleukin 2 on thymocyte activation. We have also detected in vivo the functional consequences of N-ras deficiency. KO-N-ras mice showed an increased sensitivity to influenza infection, especially when low doses of virus were used. Finally, we have detected an abnormal activation pattern of downstream Ras molecules in T-cell receptor-activated KO-N-ras thymocytes that is consistent with the defective T-cell function found in these animals. All of the results derived from this work constitute a significant contribution to the knowledge of N-ras-specific functions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genes, ras/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , ras Proteins/deficiency , Animals , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , ras Proteins/genetics , ras Proteins/immunology , ras Proteins/metabolism
11.
J Virol ; 76(7): 3145-57, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884539

ABSTRACT

Prolonged expression of a ras oncogene in primary cells accelerates the natural process of senescence. This ras-induced permanent growth arrest is bypassed in cells expressing the simian virus 40 large T antigen. Previously we showed that two regions of T antigen, a region consisting of the N-terminal 147 amino acids and a region consisting of amino acids 251 to 708 (T251-708), independently overcome ras-induced senescence. Coexpression of either T-antigen fragment and Ras results in the appearance of dense foci of transformed cells. Using a series of mutants that produce shorter T-antigen fragments, we show that the C-terminal limit of the N-terminal T-antigen fragment that cooperates with Ras lies between amino acids 83 and 121. The N-terminal limit of the C-terminal T-antigen fragment lies between amino acids 252 and 271. In addition, we present evidence that cooperation between the N-terminal fragment and Ras depends upon an intact T-antigen J domain and the ability of the T antigen to bind and inactivate the growth-suppressive effect of the tumor suppressor Rb. Introduction of specific amino acid substitutions surrounding residue 400 into T251-708 prevented the fragment from cooperating with Ras. T251-708 proteins with these same substitutions inhibited the transcriptional transactivating potential of p53 as effectively as did the wild-type protein. Thus, at least one activity contained within T251-708, other than inactivating p53 as a transcriptional transactivator, is likely to be required to bypass Ras-induced senescence.


Subject(s)
Antigens, Viral, Tumor/immunology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Genes, ras/immunology , Simian virus 40/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Cellular Senescence , Cyclin A/genetics , Fibroblasts/virology , Molecular Sequence Data , Mutation , Protein Binding , Rats , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/immunology
12.
Cancer ; 92(6): 1525-30, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11745231

ABSTRACT

BACKGROUND: The majority of lung carcinoma cases occur in current or former smokers. K-ras gene mutations are common in lung adenocarcinoma and have been associated with cigarette smoking, asbestos exposure, and female gender. METHODS: In the current study, the authors examined the contribution of cigarette smoking to K-ras gene mutations in patients with primary lung adenocarcinoma. Smoking histories were obtained from 106 prospectively enrolled patients with primary adenocarcinoma of the lung. RESULTS: K-ras mutations were detected in the primary tumor using an allele-specific ligation assay. Ninety-two of the 106 patients (87%) with lung adenocarcinoma were smokers. Nonsmokers with this tumor were more likely to be women (11 of 14; 79%), whereas the majority of smokers (57%) were men. K-ras mutations were detected in 40 of 106 tumors (38%) and were significantly more common in smokers compared with nonsmokers (43% vs. 0%; P = 0.001). CONCLUSIONS: The results of the current study confirm and extend previous observations that smokers with adenocarcinoma of the lung are more likely to have K-ras mutant tumors compared with nonsmokers. The strong link between cigarette smoking and K-ras mutations in adenocarcinoma of the lung supports the role of specific tobacco carcinogens in the etiology of this malignancy.


Subject(s)
Adenocarcinoma/immunology , Genes, ras/immunology , Lung Neoplasms/immunology , Mutation , Smoking/adverse effects , Aged , Female , Humans , Male , Prospective Studies , Sex Factors
13.
J Immunol ; 167(1): 123-31, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418640

ABSTRACT

CD44 is a widely expressed integral membrane glycoprotein that serves as a specific adhesion receptor for the extracellular matrix glycosaminoglycan hyaluronan. CD44 participates in a variety of physiological and pathological processes through its role in cell adhesion. Under appropriate conditions, the ectodomain of CD44 is proteolytically removed from the cell surface. In this study we show that excessive CD44 shedding can be induced in mouse fibroblasts and monocytes upon exposure of these cells to a CD44-specific Ab immobilized on plastic, whereas treatment with phorbol ester induces significantly enhanced CD44 release from the monocytes only. CD44 shedding proceeds normally in fibroblasts and monocytes deficient in TNF-alpha converting enzyme (TACE), a sheddase involved in the processing of several substrates. Conversely, activation of the CD44 protease has no effect on the release of TNF-alpha from TACE-expressing cells, although the same metalloprotease inhibitor effectively blocks both TACE and the CD44 sheddase. Concomitant with anti-CD44 Ab- or phorbol ester-induced CD44 shedding, dramatic changes are observed in cell morphology and the structure of the actin cytoskeleton. Disruption of actin assembly with cytochalasin reduces CD44 shedding, but not the release of TNF-alpha. Moreover, pharmacological activation of Rho family GTPases Rac1 and Cdc42, which regulate actin filament assembly into distinct cytoskeletal structures, has a profound effect on CD44 release. We conclude that the CD44 sheddase and TACE are distinct enzymes, and that Ab- and phorbol ester-enhanced cleavage of CD44 is controlled in a cell type-dependent fashion by Rho GTPases through the cytoskeleton.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cytoskeleton/immunology , Cytoskeleton/metabolism , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/metabolism , Bone Marrow Cells/immunology , Cell Adhesion/immunology , Cell Line, Transformed , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Culture Media, Conditioned , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/metabolism , Genes, myc/immunology , Genes, ras/immunology , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Hydrolysis , Kinetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/deficiency , Mice , Mice, Inbred BALB C , Monocytes/enzymology , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/biosynthesis , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Tetradecanoylphorbol Acetate/pharmacology
15.
Dig Dis Sci ; 45(12): 2290-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11258547

ABSTRACT

As an animal model for human inflammatory bowel disease and colorectal cancer, the cotton-top tamarin remains controversial. Demonstration of antigenic similarity to the human would enhance its validity. Using colonic extracts and washings, we compared binding of seven monoclonal antibodies reactive with bowel and cancer antigens in both tamarins and humans with inflammatory bowel disease. Additionally, telomerase activity was tested for. Expression of a mucin antigen specific to human cancer was increased in tamarin colonic washings as well as aminoproteoglycans and EGFR in tamarin extracts, as compared to those of humans with inflammatory bowel disease (P < 0.005). An adenoma-associated antigen and k-ras p21 protein were negative in the tamarin. A trend to greater telomerase activity exists in tamarins. The antigenic similarity validates this model for human inflammatory bowel disease and colorectal cancer. A trend to increased telomerase activity in tamarins is consistent with the greater predisposition to cancer in these animals.


Subject(s)
Antigens/analysis , Digestive System/immunology , Disease Models, Animal , Inflammatory Bowel Diseases/immunology , Saguinus/immunology , Animals , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Colorectal Neoplasms/immunology , Genes, ras/immunology , Humans , Telomerase/metabolism
16.
Scand J Immunol ; 49(4): 381-90, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10219763

ABSTRACT

Extensive evidence supports a role for natural antibody (NAb) acting against small tumour foci in vivo. Ras-transformation of murine C3H 10T 1/2 fibroblasts, known to partially activate and down-regulate endogenous PKC-alpha, increased their serum NAb-binding capacity consistent with the requirements for natural immune surveillance. Now a rat PKC-beta1-overexpressing 10T 1/2 clone, PKC-4, with an 11-fold increase in PKC activity and an activated, partially transformed phenotype, links higher susceptibility to transformation through v-Ha-ras infection with an 80% increase in NAb binding assayed by flow cytometry. H7 and E-64d inhibition and phorbol ester depletion of PKC reduced NAb binding. PKC-beta1 expression and NAb binding exhibited a similar temporal recovery from TPA treatment. Thus, expression of NAb-binding structures appears to be elevated by constitutive increases in the basal activation of PKC in both the ras-transformation and the PKC-beta1-preneoplasia models. This, coupled with corresponding decreases in membrane PKC-alpha and NAb binding in confluent 10T 1/2 cells raises the possibility that in general, cells activated through PKC are NAb sensitive. Together with the increased in vivo elimination of the high NAb-binding PKC-4 cells, the data extend the support for a role for NAb in immune surveillance, to resistance against preneoplastic cells, and argue for NAb contributing to homeostasis of the organism.


Subject(s)
Binding Sites, Antibody , Isoenzymes/biosynthesis , Precancerous Conditions/enzymology , Precancerous Conditions/immunology , Protein Kinase C/biosynthesis , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Binding Sites, Antibody/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/immunology , Genes, ras/immunology , Immunity, Innate , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C3H , Precancerous Conditions/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C beta , Rats , Tetradecanoylphorbol Acetate/pharmacology , Tumor Stem Cell Assay
17.
J Immunol ; 160(5): 2433-41, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9498787

ABSTRACT

We recently identified a murine mutant Ras p21 CD8+ CTL epitope reflecting residues 4 to 12, containing the mutation of Gly to Val at codon 12, that bound weakly to H-2Kd in vitro and generated a weak primary CTL response in immunized BALB/c mice. Here, we explored the hypothesis that specific modifications to the Ras4-12 peptide sequence can improve MHC binding, leading to enhanced immunogenicity without altering immune specificity. We synthesized Ras4-12 peptides in which Val at residue 12 was replaced with the more dominant H-2Kd C-terminus anchor residue Leu or Ile. In functional H-2Kd binding assays, Ras4-12(L12 or I12) peptide variants competed more effectively than the Ras4-12(V12) peptide. Ras4-12(L12 or I12) peptide variants enhanced both in vitro cytotoxicity and proliferation responses of anti-Ras4-12 CTL compared with the mutant Ras4-12(V12) peptide. Additionally, the Ras4-12(L12) peptide variant induced a quantitatively greater T cell response in vivo compared with that produced by Ras4-12(V12) as determined by IFN-gamma production. Mice immunized with Ras4-12(L12) peptide elicited CD8+ CTL activity specific for target cells presenting the Ras4-12(V12) epitope exogenously and endogenously. Moreover, both anti-Ras4-12(V12)-derived and anti-Ras4-12(L12)-derived CTL lines were similar insofar as their TCR usage and amino acid contact residues in the Ras4-12(V12) peptide. These experiments demonstrate that modifications can be introduced in tumor-specific peptide epitopes to enhance both in vitro and in vivo immunogenicity. The design of oncogene-specific peptide epitope variants as immunogens may accelerate the generation of anti-tumor T cell responses for cancer immunotherapy.


Subject(s)
Epitopes/immunology , H-2 Antigens/metabolism , Mutagenesis , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/metabolism , ras Proteins/genetics , ras Proteins/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , CD8 Antigens/physiology , Cell Line , Cytotoxicity, Immunologic/drug effects , Epitope Mapping , Epitopes/genetics , Epitopes/metabolism , Female , Genes, ras/immunology , H-2 Antigens/genetics , H-2 Antigens/physiology , Injections, Subcutaneous , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes, Cytotoxic/immunology , ras Proteins/administration & dosage , ras Proteins/metabolism
19.
J Exp Clin Cancer Res ; 16(1): 15-22, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9148855

ABSTRACT

Strong immunogenicity is induced by antitumor triazene compounds in tumor cells through a mutagenic mechanism. A highly immunogenic <> clone, isolated from a dacarbazine-treated L5178Y leukemia of DBA/2 mice, was transfected with K-ras mutated at codon 12 (i.e. ras(m12)). This transfected clone presents at least 2 mutations, one concerning K-ras gene, and the other affecting an unrelated gene, responsible for the generation of a highly immunogenic, MHC class I restricted non-self peptide. The results indicate that cells of <> clone transfected with ras(m12) were less immunogenic than cells of the same origin transfected with the vector alone. Moreover, ras(m12)-transfected cells showed lower levels of H-2K(d) gene expression with respect to those detectable in control cells. In addition, in vivo and in vitro sensitization against <> clone carrying mutated ras did not result in a strong cytotoxic T lymphocyte response against ras(m12)-transfected non immunogenic L5178y target cells. These preliminary results suggest that K-ras mutation could down-regulate the level of tumor immunogenicity, possibly acquired through a mutagenic process affecting other unrelated genes.


Subject(s)
Antineoplastic Agents/pharmacology , Dacarbazine/pharmacology , Genes, ras/immunology , Leukemia L5178/immunology , Transfection , Animals , Genes, ras/genetics , Histocompatibility Antigens Class I/analysis , Leukemia L5178/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Transfection/genetics
20.
Zhonghua Yi Xue Za Zhi ; 76(9): 650-3, 1996 Sep.
Article in Chinese | MEDLINE | ID: mdl-9275544

ABSTRACT

OBJECTIVE: To investigate if treatment with antisense H-ras oligodeoxynucleotides (ODNs) modulates tumor growth, apoptosis and metastasis of a high metastatic tumor model of human hepatocellular carcinoma (HCC) in nude mice LCI-D20, in which over-expression of H-ras has been identified. METHODS: LCI-D20 cells in primary culture were treated with 10 mumol/L antisense ODNs in vitro. 1.5 x 10(6) LCI-D20 cells with or without pretreatment were inoculated into each elevated subcutaneous (s.c) flap in fourteen nude mice, 6 animals for antisense H-ras ODN treated cells, 4 for H-ras non-specific antisense ODN treated cells, the rest 4 for cells without pretreatment. RESULTS: In in vitro cell culture study, 5-day continuous suppression of H-ras expression by antisense H-ras ODNs resulted in significant inhibition of the proliferation of LCI-D20 cells (t = 31.529, P < 0.01). Cell cycle analysis showed a significant decrease in S phase (36.0 +/- 1.4) and a remarkable increase in G1/G0 fraction (56.7 +/- 1.1) after exposure to antisense H-ras ODNs in comparison with the cells without any treatment (58.5 +/- 0.9, t = 13.519, P < 0.01, 37.4 +/- 0.7, t = 14.802, P < 0.01). In situ end-labeling (ISEL) detection showed that apoptotic cell death was significantly increased in cells with 5-day treatment of antisense H-ras ODNs (34.0% +/- 4.5%) in comparing with cells without treatment (2.5% +/- 1.2%, t = 13. 434, P < 0.01) or treated with non-specific antisense ODNs (4.8% +/- 1.4%, t = 12.453, P < 0.01) at the corresponding time. In in vivo experiment, after six-week observation, tumor growth in antisense H-ras treated animals was significantly retarded in comparison with that of the untreated (t = 3.509, P < 0.01) or nonspecific antisense ODN treated animals (t = 3.452, P < 0.01). CONCLUSIONS: Specific inhibition of H-ras expression by antisense H-ras ODNs could not only induce apoptotic cell death, inhibit the growth rate of LCI-D20 cells in vitro and in vivo, but also alter in vivo tumorigenesity and metastatic potential of LCI-D20 cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Genes, ras/immunology , Liver Neoplasms/pathology , Oligonucleotides, Antisense , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...