Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Lab Invest ; 81(2): 201-10, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11232642

ABSTRACT

The deleted in colorectal cancer (DCC) protein is important in the pathway guidance of cells and cell processes during neural development, and DCC has also been implicated in the aberrant cellular migrations of neuroblastoma dissemination. We attempted to further define DCC protein function by the overexpression of full-length and truncated DCC constructs in a human neuroblastoma cell line. Overexpression of the truncated DCC protein resulted in a less epithelioid morphology. This was accompanied by decreases in expression of N-cadherin and alpha- and beta-catenin by immunoblot and Northern blot analysis. Levels of desmoglein were relatively less affected, whereas endogenous DCC protein levels were increased in the truncated transfectants. N-cadherin immunofluorescence was consistent with the immunoblot studies and localized the protein to the cytoplasm and sites of cell-cell contact. Cell aggregation studies demonstrated diminished calcium-dependent aggregation in the truncated transfectants. In conclusion, overexpression of a truncated DCC protein in neuroblastoma cells resulted in the loss of an epithelioid morphology, diminished expression of N-cadherin and alpha- and beta-catenin, and diminished calcium-dependent cell adhesion. These studies provide the first evidence of an apparent functional link between DCC and N-cadherin/catenin-dependent cell adhesion.


Subject(s)
Cadherins/genetics , Calcium/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Trans-Activators , Tumor Suppressor Proteins , Cell Aggregation , Colorectal Neoplasms/genetics , Cytoskeletal Proteins/analysis , DCC Receptor , Desmogleins , Desmoplakins , Genes, DCC , Humans , Neuroblastoma , Receptors, Cell Surface , Recombinant Proteins/metabolism , Sequence Deletion , Transfection , Tumor Cells, Cultured , alpha Catenin , beta Catenin
2.
Lab Invest ; 78(6): 669-75, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9645757

ABSTRACT

Neuroblastoma is an embryonal tumor of neural crest origin noted for its heterogeneity at the clinical, histologic, and molecular levels. The deleted in colorectal cancer (DCC) protein is an adhesion family molecule of unequivocal importance in neural development that has also been implicated in several malignancies, including neuroblastoma, through its apparent loss of function. Immunohistochemical assessment of the DCC protein was performed on a group of 49 neuroblastoma specimens and examined in relation to important clinical, histologic, and molecular parameters. DCC expression was significantly associated with neuroblastoma dissemination as primary tumors from Stage 1 to 3 patients (15/20, 75%) more frequently exhibited the DCC protein than those from Stage 4 patients (5/13, 38%; p = 0.0415). Primary tumors were more frequently DCC-positive (20/33, 61%) as compared with metastatic deposits (3/16, 19%; p = 0.0063), and a single case of a paired primary and metastatic deposit demonstrated the apparent loss of DCC gene expression with tumor progression. The remaining five paired specimens were DCC-negative in both the primary tumor and metastatic deposit. No significant association was appreciated between DCC expression and patient age, the Shimada histologic classification, or N-Myc amplification. These results provide evidence that DCC expression may be lost in the course of metastatic spread in a subset of neuroblastomas. Moreover, DCC function is implicated in neuroblastoma dissemination in a manner independent of N-Myc.


Subject(s)
Gene Expression/physiology , Genes, DCC/genetics , Neuroblastoma/genetics , Neuroblastoma/secondary , Tumor Suppressor Proteins , Cell Adhesion Molecules/metabolism , DCC Receptor , Disease Progression , Humans , Immunohistochemistry , Neoplasm Staging , Neuroblastoma/pathology , Receptors, Cell Surface
3.
Cancer Res ; 57(3): 382-6, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012460

ABSTRACT

The deleted in colorectal cancer (DCC) gene, a candidate tumor suppressor gene on chromosome 18q21, encodes a neural cell adhesion molecule family protein that is most highly expressed in the nervous system. To address the hypothesis that DCC may play a role in glioma development and/or progression, we examined DCC expression by immunohistochemistry in 57 resected human astrocytic tumors. Overall, low-grade astrocytomas were predominantly DCC positive (15 of 16, or 94%), whereas high-grade tumors significantly less often expressed the DCC protein (27 of 41, or 66%; P = 0.03). We were able to directly assess the relationship between DCC expression and tumor progression in 15 patients who initially presented with a low-grade astrocytoma and subsequently recurred with a glioblastoma. Within this panel of paired lesions from the same patient, 14 of 15 (93%) low-grade tumors expressed the DCC protein, whereas only 7 of 15 (47%) corresponding glioblastomas were DCC positive. We also observed that secondary glioblastomas resulting from malignant progression of low-grade astrocytomas were more often DCC negative (8 of 15, or 53%) compared with primary or de novo glioblastomas (6 of 26, or 23%; P = 0.05). These findings implicate DCC inactivation in glioma progression and also demonstrate that DCC expression is preferentially, but not exclusively, lost in the genetic pathway to secondary glioblastoma multiforme.


Subject(s)
Genes, DCC , Glioma/genetics , Animals , Astrocytes/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioma/pathology , Humans , Mice
4.
N Engl J Med ; 335(23): 1727-32, 1996 Dec 05.
Article in English | MEDLINE | ID: mdl-8929264

ABSTRACT

BACKGROUND: Allelic loss of chromosome 18q predicts a poor outcome in patients with stage II colorectal cancer. Although the specific gene inactivated by this allelic loss has not been elucidated, the DCC (deleted in colorectal cancer) gene is a candidate. We investigated whether the expression of the DCC protein in tumor cells is a prognostic marker in colorectal carcinoma. METHODS: The expression of DCC was evaluated immunohistochemically in 132 paraffin-embedded samples from patients with curatively resected stage II and III colorectal carcinomas. The Cox proportional-hazards model was used to adjust for covariates including age, sex, tumor site, degree of tumor differentiation, and use of adjuvant therapy. RESULTS: The expression of DCC was a strong positive predictive factor for survival in both stage II and stage III colorectal carcinomas. In patients with stage II disease whose tumors expressed DCC, the five-year survival rate was 94.3 percent, whereas in patients with DCC-negative tumors, the survival rate was 61.6 percent (P<0.001). In patients with stage III disease, the respective survival rates were 59.3 percent and 33.2 percent (P=0.03). CONCLUSIONS: DCC is a prognostic marker in patients with stage II or stage III colorectal cancer. In stage II colorectal carcinomas, the absence of DCC identifies a subgroup of patients with lesions that behave like stage III cancers. These findings may thus have therapeutic implications in this group of patients.


Subject(s)
Biomarkers, Tumor/analysis , Cell Adhesion Molecules/analysis , Colorectal Neoplasms/chemistry , Genes, DCC/genetics , Tumor Suppressor Proteins , Aged , Biomarkers, Tumor/genetics , Cell Adhesion Molecules/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DCC Receptor , Female , Gene Expression , Humans , Immunohistochemistry , Life Tables , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Receptors, Cell Surface , Survival Rate
5.
Clin Cancer Res ; 2(7): 1097-102, 1996 Jul.
Article in English | MEDLINE | ID: mdl-9816273

ABSTRACT

DCC, a candidate tumor suppressor gene from chromosome 18q21, is most highly expressed in the developing nervous system. In vitro studies suggest a role for DCC in neuronal differentiation, and 18q allelic loss occurs in a subset of neuroblastomas. To address the hypothesis that loss of DCC function may contribute to tumorigenesis in cells of neural origin, we utilized a combination of RNase protection, immunoblotting, and immunohistochemical approaches to characterize DCC expression in 62 primary neuroblastomas and 16 neuroblastoma cell lines. The DCC protein was undetectable in 38% of the primary tumors and 56% of the cell lines. Of note, primary tumors lacking DCC expression were more likely to have been obtained from patients with disseminated or stage D disease (P = 0.01). In addition, loss of DCC expression was observed in three of six primary tumors from stage DS patients. No consistent relationship between the loss of DCC expression and N-myc amplification was observed in our studies. Our findings suggest that loss of DCC expression may contribute to the dissemination of neuroblastoma cells, perhaps through alterations in growth and differentiation pathways distinct from those regulated by N-myc.


Subject(s)
Genes, DCC , Neuroblastoma/genetics , Tumor Suppressor Proteins , Cell Adhesion Molecules/analysis , DCC Receptor , Genes, myc , Humans , Immunoblotting , Immunohistochemistry , Neuroblastoma/pathology , Receptors, Cell Surface , Tumor Cells, Cultured
6.
South Med J ; 88(4): 462-4, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7536347

ABSTRACT

Therapy with myeloid colony-stimulating factors has been safely and effectively used in a wide variety of situations associated with neutropenia. We present a case of pseudoleukemia occurring in a patient with lymphoma and pancytopenia after 2 days of treatment with granulocyte colony-stimulating factor (G-CSF). Bone marrow aspirate and flow cytometry study results were consistent with acute myelomonocytic leukemia but were normal after G-CSF was discontinued for 4 days. As previous phase I studies of bone marrow morphology after G-CSF use have not described the extreme myeloid immaturity seen in this patient, it seems likely that the action of G-CSF was enhanced by factors associated with the patient's illness. We emphasize the clinical importance of this case in light of the widespread use of G-CSF.


Subject(s)
Granulocyte Colony-Stimulating Factor/adverse effects , Leukemia, Myelomonocytic, Acute/etiology , Neoplasms, Second Primary/etiology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Humans , Leukemia, Myelomonocytic, Acute/pathology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Neutropenia/therapy , Prednisone/administration & dosage , Vincristine/administration & dosage
7.
Dev Biol ; 166(2): 654-65, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7813784

ABSTRACT

The deleted in colorectal cancer (DCC) gene has been identified as a candidate tumor suppressor gene on the basis of frequent allelic loss and decreased or absent gene expression in several human cancer types, as well as somatic mutations in the gene in colorectal tumors. We have identified a Xenopus DCC homologue (XDCC alpha) predicted to encode a protein of 1427 amino acids and have characterized XDCC expression in developing embryos and adult tissues. The predicted amino acid sequences of XDCC alpha and human DCC are greater than 80% identical; each has four immunoglobulin-like domains, six fibronectin type III domains, and a cytoplasmic domain of about 325 amino acids. While RNase protection assays and immunoblotting studies failed to detect XDCC alpha expression in embryos prior to developmental stage 15, XDCC alpha expression was present in embryos from stages 19 to 46. Whole mount in situ hybridization studies localized XDCC alpha expression to developing forebrain, midbrain, and hindbrain regions. DCC expression was inhibited by treatments that altered the development of mature neural structures; specifically, uv-ventralized embryos and exogastrulae had reduced DCC expression. These results indicate that XDCC alpha is developmentally regulated and expressed as a consequence of neural induction. Moreover, unlike some well-characterized tumor suppressor genes, such as the p53 and retinoblastoma genes, that are not differentially expressed in developing Xenopus embryos, the DCC gene may have a specific role in the morphogenesis of the brain and perhaps other tissues and organs.


Subject(s)
Cell Adhesion Molecules/genetics , Genes, DCC , Nervous System/embryology , Tumor Suppressor Proteins , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Cloning, Molecular , DCC Receptor , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization , Molecular Sequence Data , RNA, Messenger/genetics , Receptors, Cell Surface , Sequence Alignment , Sequence Homology, Amino Acid , Xenopus laevis/genetics
8.
Cancer Res ; 54(16): 4493-501, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-8044801

ABSTRACT

The DCC (deleted in colorectal cancer) gene was identified because it is affected by somatic mutations in colorectal tumors, including allelic losses in greater than 70% of cancers and localized mutations in a subset of cases. The DCC gene also may be inactivated in other tumor types, including cancers of the pancreas, stomach, breast, prostate, and brain, as well as some leukemias. We have characterized DCC complementary DNAs obtained from human fetal brain tissues and IMR32 human neuroblastoma cells. Based on the fetal brain complementary DNA sequence, the predicted transmembrane DCC protein product has 1447 amino acids. The extracellular domain of about 1100 amino acids has four immunoglobulin-like domains and six fibronectin type III-like domains; the 325-amino acid cytoplasmic domain does not show similarity to previously characterized proteins. Comparison of DCC complementary DNAs from IMR32 cells to those from fetal brain identified two potential alternative splice sites. Studies of adult mouse tissues revealed that DCC transcripts were present at very low levels in all tissues studied, and alternative splicing of DCC transcripts was seen in some tissues. Immunoblotting and immunoprecipitation studies with DCC-specific antisera identified protein species with molecular weights of approximately 175,000-190,000 in some rodent tissues and human tumor cell lines. DCC protein expression was highest in brain tissues and neural crest-derived cell lines and markedly reduced or absent in the majority of cancer cell lines studied. Treatment of DCC-expressing cells with tunicamycin decreased the apparent molecular weight of the immunoreactive proteins, establishing that DCC is a glycoprotein. The studies presented here demonstrate that the DCC gene encodes several related glycoprotein species that are likely to be expressed at very low levels in many normal adult tissues. Furthermore, the absence of DCC expression in some of the cancer cell lines studied may result from genetic inactivation of DCC.


Subject(s)
Alternative Splicing/genetics , Brain , Colon , Gene Expression Regulation, Neoplastic/genetics , Genes, DCC/genetics , Neuroblastoma/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Brain Chemistry , Chromosome Mapping , Colon/chemistry , DNA, Complementary/genetics , Exons/genetics , Fetus , Gene Deletion , Humans , Mice , Molecular Sequence Data , Molecular Weight , Neoplasm Proteins/analysis , Neuroblastoma/chemistry , Polymerase Chain Reaction , Rats , Sequence Analysis, DNA , Transfection , Tumor Cells, Cultured
9.
J Cell Biol ; 124(6): 1017-27, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8132705

ABSTRACT

The Deleted in Colorectal Cancer (DCC) gene is a candidate tumor suppressor gene that is predicted to encode a transmembrane polypeptide with strong similarity to the neural cell adhesion molecule (N-CAM) family. Previous studies have suggested that several different N-CAMs, when expressed in non-neuronal cell types can stimulate neurite outgrowth from PC12 rat pheochromocytoma cells. Based on the predicted structural similarity of DCC to N-CAMs, we sought to determine whether NIH3T3 cells expressing DCC could stimulate neurite outgrowth in PC12 cells. We found that NIH3T3 cell lines expressing DCC could stimulate PC12 cells to extend neurites. Supernatants from DCC-transfected NIH3T3 cells did not induce neurite outgrowth above background levels, suggesting that cell-cell interaction was required. NIH3T3 cells expressing a truncated form of DCC, lacking the majority of the cytoplasmic domain sequences, also failed to induce neurite outgrowth above the levels seen with control NIH3T3 cells, suggesting that the cytoplasmic domain of DCC was necessary for its neurite-promoting function. In contrast to NGF-mediated neurite outgrowth, the DCC-mediated response was inhibited by treatment with pertussis toxin or the combination of N- and L-type calcium channel blockers, and was unaffected by the transcriptional inhibitor cordycepin. The data suggest that the DCC protein can function in a fashion analogous to other N-CAMs to alter PC12 cell phenotype through intracellular pathways distinct from those involved in NGF signaling.


Subject(s)
Genes, DCC , Membrane Proteins/physiology , Neurites/physiology , 3T3 Cells , Animals , Calcium Channel Blockers/pharmacology , Cell Differentiation , Cell Membrane/chemistry , Deoxyadenosines/pharmacology , Diltiazem/pharmacology , Fluorescent Antibody Technique , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , PC12 Cells , Peptides/pharmacology , Pertussis Toxin , RNA, Messenger/biosynthesis , Rats , Transfection , Virulence Factors, Bordetella/pharmacology , omega-Conotoxin GVIA
10.
Article in English | MEDLINE | ID: mdl-7587124

ABSTRACT

Chromosome 18q is among the chromosomal regions thought to harbor a tumor suppressor gene(s) that is frequently inactivated during the development of several cancer types, particularly those of the gastrointestinal tract. Moreover, preliminary data suggest that colorectal cancers with 18q LOH have a more aggressive clinical behavior than those cancers without 18q LOH. A candidate tumor suppressor gene from 18q, termed DCC for deleted in colorectal cancer, has been identified. The DCC gene is contained within the common region of LOH on 18q, its expression is markedly decreased or absent in colorectal cancers and cell lines, and a subset of colorectal cancers have been shown to have somatic mutations within the DCC gene. Thus, DCC represents the most promising candidate tumor suppressor gene from 18q. At present, however, many questions remain regarding the mechanisms underlying the inactivation of DCC in the majority of colorectal cancers. In addition, although studies of 18q LOH and DCC gene expression in other cancer types suggest that DCC inactivation may contribute to the pathogenesis of other tumor types, few studies have provided definitive data to demonstrate that DCC inactivation is a critical genetic event in these tumors. Moreover, little is known about the function of DCC in the regulation of normal cell growth and tumor suppression. The predicted structural similarity of DCC to the N-CAM family of cell-surface proteins suggests that it may function through cell-cell and/or cell-extracellular matrix interactions.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Colorectal Neoplasms/genetics , Genes, DCC , Tumor Suppressor Proteins , Animals , Base Sequence , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Differentiation/genetics , DCC Receptor , DNA Primers/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Molecular Sequence Data , Neoplasms/genetics , Polymerase Chain Reaction , Receptors, Cell Surface , Tissue Distribution
11.
J Immunol ; 139(6): 1985-90, 1987 Sep 15.
Article in English | MEDLINE | ID: mdl-3114381

ABSTRACT

We have dissected the human influenza-specific B cell repertoire by performing Epstein-Barr virus (EBV) limiting dilution analysis of lymphocytes obtained from donors before and after immunization with a commercially available influenza vaccine. In addition to an analysis of precursor frequency and light chain diversity, we studied sera and culture supernatants containing human anti-influenza antibodies with a panel of murine monoclonal antibodies specific for idiotopes identified on murine anti-PR8 and anti-X-31 antibodies. An idiotypic specificity present on the X-31-specific murine monoclonal PY206 has previously been shown to be shared by murine antibodies specific for PR8, X-31, and other influenza viruses. We observed little correlation among the following parameters: anti-viral titer, serum idiotope content, precursor frequency and immune status. More interestingly, there was a striking predominance of human influenza-specific antibodies that utilized lambda light chains. In addition, 12 of 26 human anti-influenza monoclonals strongly inhibited the binding of one of the murine anti-idiotopes to the labeled murine antibody, PY206. This is the same idiotope that is shared among murine antiinfluenza antibodies and all six individuals studied contained clones reactive with this anti-idiotope. Seven of these 12 idiotope-positive human antibodies gave partial cross-reactivity in a second anti-idiotypic system. These observations imply that a significant level of homology exists between the binding sites of human and murine influenza-specific antibodies and suggest that idiotypic manipulation of the human immune response to influenza virus may have important therapeutic implications.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Immunoglobulin Idiotypes/immunology , Orthomyxoviridae/immunology , Adult , Animals , Hemagglutinins, Viral/immunology , Humans , Immunoglobulin Light Chains/analysis , Mice , Middle Aged , Neutralization Tests , Species Specificity , Viral Vaccines/immunology
12.
Viral Immunol ; 1(1): 1-12, 1987.
Article in English | MEDLINE | ID: mdl-3509671

ABSTRACT

Several syngeneic monoclonal anti-idiotypic antibodies were obtained against PY206, a monoclonal antibody specific for X-31 (H3N2) influenza virus hemagglutinin. This idiotype was found in the sera of BALB/c mice immunized with various influenza viruses. Adsorption experiments indicated that the PY206 Id was borne by antibodies specific for viral hemagglutinin (HA) and/or neuraminidase (NA). This idiotype was identified on other monoclonal antibodies specific for various influenza HAs (H3 and H1). Study of the variable-region (V) genes of these monoclonal antibodies showed that its expression is independent of variable kappa (VK)21 light-chains and that the heavy-chains of the strongly idiotype-positive hybridomas derive from either the variable heavy (VH) J558 or VH 7183 family. Finally, Western blot analysis demonstrated that PY206 idiotypic determinants are located exclusively on the heavy chain.


Subject(s)
Genes, Immunoglobulin , Immunoglobulin Idiotypes/analysis , Orthomyxoviridae/immunology , Animals , Antibodies, Monoclonal , Antigens, Viral/immunology , Blotting, Western , Cross Reactions , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Hemagglutinins, Viral/immunology , Hybridomas , Immunoglobulin Variable Region/genetics , Mice , Mice, Inbred BALB C , Radioimmunoassay
14.
J Immunol ; 137(4): 1352-8, 1986 Aug 15.
Article in English | MEDLINE | ID: mdl-2426361

ABSTRACT

A panel of monoclonal antibodies specific for a corresponding panel of sequentially selected variants of influenza A/PR/8/34 virus has been established. Although the monoclonal antibodies are paratypically distinct, idiotypic relatedness has been observed. Two cross-reactive idiotypes have been defined that are associated with the 7183 and S107 VH gene families, respectively. Three of the four monoclonal antibodies utilize the VK21 group of light chains, and three VH genes belong to the VH7183 family and one to the VH S107 family. Antibodies encoded by genes deriving from the VH7183 family share a cross-reactive idiotype, a marker of the VH region as well as distinct individual idiotopes. These antibodies are produced by different clones using related VH and VK genes.


Subject(s)
Antibodies, Monoclonal/analysis , Antibodies, Viral/analysis , Antibody Specificity , Influenza A virus/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Epitopes/immunology , Genetic Variation , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Idiotypes/genetics , Immunoglobulin Idiotypes/immunology , Immunoglobulin Variable Region/genetics , Influenza A virus/genetics , Mice , Mice, Inbred BALB C , Rabbits
16.
J Virol ; 53(2): 425-9, 1985 Feb.
Article in English | MEDLINE | ID: mdl-3871483

ABSTRACT

The humoral response to influenza A/PR8 virus was examined in the CBA/N and C3J.xid strains of mice, both of which bear an X-linked genetic defect (xid), and in strains lacking this defect. Hemagglutination-inhibiting antibody titers and measurement of virus-specific antibodies by solid-phase radioimmunoassay indicated that the xid defect does not impair the production of an adequate anti-influenza antibody response. However, investigation of the isotypes of PR8 virus-specific antibodies disclosed a relative decrease in the levels of IgG3 and IgG1 in the xid-bearing strains. This was observed after both intraperitoneal immunization and aerosol infection. The isotype differences were not reflected in the susceptibility of these strains to influenza virus infection.


Subject(s)
Antibodies, Viral/biosynthesis , B-Lymphocytes/immunology , Immunoglobulin Allotypes/biosynthesis , Immunoglobulin G/biosynthesis , Immunologic Deficiency Syndromes/immunology , Influenza A virus/immunology , Animals , Hemagglutination Inhibition Tests , Immunization , Immunoglobulin M/biosynthesis , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Orthomyxoviridae Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...