Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Magn Reson Med ; 72(5): 1408-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24281951

ABSTRACT

PURPOSE: A practical, noninvasive method is needed to measure the extracellular pH (pHe) within in vivo tumors to longitudinally monitor tumor acidosis. We have optimized a biomedical imaging method, termed acidoCEST MRI, to provide noninvasive assessments of tumor pHe in preclinical models of mammary carcinoma. METHODS: A CEST-FISP MRI method was optimized to detect the chemical exchange saturation transfer (CEST) of two amide protons of a clinically approved CT contrast agent, iopromide. The ratio of the two CEST effects was used to measure pH. Routes of administration of iopromide were evaluated to ensure sufficient delivery of the agent to the tumor. The optimized acidoCEST MRI method was then used to evaluate the change in tumor pHe following alkalinizing bicarbonate treatment. RESULTS: The acidoCEST MRI protocol measured pH between 6.2 and 7.2 pH units. Greater delivery of iopromide was shown to improve the precision of the measurement of tumor pHe, but the agent did not influence the tumor pHe. AcidoCEST MRI was used to longitudinally monitor the effect of bicarbonate treatment on the pHe of tumors and bladders. CONCLUSION: This study demonstrates that an optimized acidoCEST MRI method is a practical, noninvasive method for assessing changes in tumor acidosis.


Subject(s)
Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/chemistry , Acidosis/diagnosis , Animals , Bicarbonates/pharmacology , Contrast Media/administration & dosage , Contrast Media/chemistry , Hydrogen-Ion Concentration , Iohexol/administration & dosage , Iohexol/analogs & derivatives , Iohexol/chemistry , Mice , X-Ray Microtomography
2.
Biomed Res Int ; 2013: 485196, 2013.
Article in English | MEDLINE | ID: mdl-23936808

ABSTRACT

The extracellular pH (pHe) of many solid tumors is acidic as a result of glycolytic metabolism and poor perfusion. Acidity promotes invasion and enhances metastatic potential. Tumor acidity can be buffered by systemic administration of an alkaline agent such as sodium bicarbonate. Tumor-bearing mice maintained on sodium bicarbonate drinking water exhibit fewer metastases and survive longer than untreated controls. We predict this effect is due to inhibition of tumor invasion. Reducing tumor invasion should result in fewer circulating tumor cells (CTCs). We report that bicarbonate-treated MDA-MB-231 tumor-bearing mice exhibited significantly lower numbers of CTCs than untreated mice (P < 0.01). Tumor pHe buffering may reduce optimal conditions for enzymes involved in tumor invasion such as cathepsins and matrix metalloproteases (MMPs). To address this, we tested the effect of transient alkalinization on cathepsin and MMP activity using enzyme activatable fluorescence agents in mice bearing MDA-MB-231 mammary xenografts. Transient alkalinization significantly reduced the fluorescent signal of protease-specific activatable agents in vivo (P ≤ 0.003). Alkalinization, however, did not affect expression of carbonic anhydrase IX (CAIX). The findings suggest a possible mechanism in a live model system for breast cancer where systemic alkalinization slows the rate of invasion.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplasm Invasiveness/pathology , Sodium Bicarbonate/administration & dosage , Animals , Breast Neoplasms/drug therapy , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Hydrogen-Ion Concentration , Mice , Neoplastic Cells, Circulating/drug effects , Xenograft Model Antitumor Assays
3.
Proc SPIE Int Soc Opt Eng ; 85742013 Feb 02.
Article in English | MEDLINE | ID: mdl-27688523

ABSTRACT

Upregulate levels of expression and activity of membrane H+ ion pumps in cancer cells drives the extracellular pH (pHe,) to values lower than normal. Furthermore, disregulated pH is indicative of the changes in glycolytic metabolism in tumor cells and has been shown to facilitate extracellular tissue remodeling during metastasis Therefore, measurement of pHe could be a useful cancer biomarker for diagnostic and therapy monitoring evaluation. Multimodality in-vivo imaging of pHe in tumorous tissue in a mouse dorsal skin fold window chamber (DSFWC) model is described. A custom-made plastic window chamber structure was developed that is compatible with both imaging optical and MR imaging modalities and provides a model system for continuous study of the same tissue microenvironment on multiple imaging platforms over a 3-week period. For optical imaging of pHe, SNARF-1 carboxylic acid is injected intravenously into a SCID mouse with an implanted tumor. A ratiometric measurement of the fluorescence signal captured on a confocal microscope reveals the pHe of the tissue visible within the window chamber. This imaging method was used in a preliminary study to evaluate sodium bicarbonate as a potential drug treatment to reverse tissue acidosis. For MR imaging of pHe the chemical exchange saturation transfer (CEST) was used as an alternative way of measuring pHe in a DSFWC model. ULTRAVIST®, a FDA approved x-ray/CT contrast agent has been shown to have a CEST effect that is pH dependent. A ratiometric analysis of water saturation at 5.6 and 4.2 ppm chemical shift provides a means to estimate the local pHe.

4.
BMC Cancer ; 11: 235, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663677

ABSTRACT

BACKGROUND: The glycolytic nature of malignant tumors contributes to high levels of extracellular acidity in the tumor microenvironment. Tumor acidity is a driving force in invasion and metastases. Recently, it has been shown that buffering of extracellular acidity through systemic administration of oral bicarbonate can inhibit the spread of metastases in a mouse model for metastatic breast cancer. While these findings are compelling, recent assessments into the use of oral bicarbonate as a cancer intervention reveal limitations. METHODS: We posited that safety and efficacy of bicarbonate could be enhanced by dichloroacetate (DCA), a drug that selectively targets tumor cells and reduces extracellular acidity through inhibition of glycolysis. Using our mouse model for metastatic breast cancer (MDA-MB-231), we designed an interventional survival study where tumor bearing mice received bicarbonate, DCA, or DCA-bicarbonate (DB) therapies chronically. RESULTS: Dichloroacetate alone or in combination with bicarbonate did not increase systemic alkalosis in mice. Survival was longest in mice administered bicarbonate-based therapies. Primary tumor re-occurrence after surgeries is associated with survival rates. Although DB therapy did not significantly enhance oral bicarbonate, we did observe reduced pulmonary lesion diameters in this cohort. The DCA monotherapy was not effective in reducing tumor size or metastases or improving survival time. We provide in vitro evidence to suggest this outcome may be a function of hypoxia in the tumor microenvironment. CONCLUSIONS: DB combination therapy did not appear to enhance the effect of chronic oral bicarbonate. The anti-tumor effect of DCA may be dependent on the cancer model. Our studies suggest DCA efficacy is unpredictable as a cancer therapy and further studies are necessary to determine the role of this agent in the tumor microenvironment.


Subject(s)
Antineoplastic Agents/therapeutic use , Bicarbonates/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dichloroacetic Acid/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Bicarbonates/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/surgery , Cell Hypoxia , Cell Line, Tumor , Dichloroacetic Acid/pharmacology , Disease Models, Animal , Female , Humans , Hydrogen-Ion Concentration , Lactates/metabolism , Lung Neoplasms/pathology , Mice , Mice, SCID , Sodium Bicarbonate/pharmacology , Sodium Bicarbonate/therapeutic use , Survival Analysis , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
5.
Math Biosci ; 230(1): 1-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21167185

ABSTRACT

Malignant tumours are characterised by a low, acidic extracellular pH (pHe) which facilitates invasion and metastasis. Previous research has proposed the potential benefits of manipulating systemic pHe, and recent experiments have highlighted the potential for buffer therapy to raise tumour pHe, prevent metastases, and prolong survival in laboratory mice. To examine the physiological regulation of tumour buffering and investigate how perturbations of the buffering system (via metabolic/respiratory disorders or changes in parameters) can alter tumour and blood pHe, we develop a simple compartmentalised ordinary differential equation model of pHe regulation by the HCO3-/CO2 buffering system. An approximate analytical solution is constructed and used to carry out a sensitivity analysis, where we identify key parameters that regulate tumour pHe in both humans and mice. From this analysis, we suggest promising alternative and combination therapies, and identify specific patient groups which may show an enhanced response to buffer therapy. In addition, numerical simulations are performed, validating the model against well-known metabolic/respiratory disorders and predicting how these disorders could change tumour pHe.


Subject(s)
Acid-Base Equilibrium/physiology , Bicarbonates/metabolism , Carbon Dioxide/metabolism , Extracellular Space/metabolism , Models, Biological , Neoplasms/metabolism , Acidosis/blood , Acidosis/complications , Acidosis/metabolism , Acidosis, Respiratory/blood , Acidosis, Respiratory/complications , Acidosis, Respiratory/metabolism , Algorithms , Alkalosis/blood , Alkalosis/complications , Alkalosis/metabolism , Alkalosis, Respiratory/blood , Alkalosis, Respiratory/complications , Alkalosis, Respiratory/metabolism , Animals , Bicarbonates/blood , Blood/metabolism , Buffers , Carbon Dioxide/blood , Computer Simulation , Humans , Hydrogen-Ion Concentration , Kinetics , Mice , Neoplasms/blood , Neoplasms/complications , Neoplasms/therapy
6.
Cancer Res ; 69(6): 2260-8, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276390

ABSTRACT

The external pH of solid tumors is acidic as a consequence of increased metabolism of glucose and poor perfusion. Acid pH has been shown to stimulate tumor cell invasion and metastasis in vitro and in cells before tail vein injection in vivo. The present study investigates whether inhibition of this tumor acidity will reduce the incidence of in vivo metastases. Here, we show that oral NaHCO(3) selectively increased the pH of tumors and reduced the formation of spontaneous metastases in mouse models of metastatic breast cancer. This treatment regimen was shown to significantly increase the extracellular pH, but not the intracellular pH, of tumors by (31)P magnetic resonance spectroscopy and the export of acid from growing tumors by fluorescence microscopy of tumors grown in window chambers. NaHCO(3) therapy also reduced the rate of lymph node involvement, yet did not affect the levels of circulating tumor cells, suggesting that reduced organ metastases were not due to increased intravasation. In contrast, NaHCO(3) therapy significantly reduced the formation of hepatic metastases following intrasplenic injection, suggesting that it did inhibit extravasation and colonization. In tail vein injections of alternative cancer models, bicarbonate had mixed results, inhibiting the formation of metastases from PC3M prostate cancer cells, but not those of B16 melanoma. Although the mechanism of this therapy is not known with certainty, low pH was shown to increase the release of active cathepsin B, an important matrix remodeling protease.


Subject(s)
Breast Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Prostatic Neoplasms/drug therapy , Sodium Bicarbonate/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
7.
Neoplasia ; 10(8): 745-56, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18670636

ABSTRACT

Malignancy in cancer is associated with aerobic glycolysis (Warburg effect) evidenced by increased trapping of [(18)F]deoxyglucose (FdG) in patients imaged by positron emission tomography (PET). [(18)F]deoxyglucose uptake correlates with glucose transporter (GLUT-1) expression, which can be regulated by hypoxia-inducible factor 1 alpha (HIF-1alpha). We have previously reported in established breast lines that HIF-1alpha levels in the presence of oxygen leads to the Warburg effect. However, glycolysis and GLUT-1 can also be induced independent of HIF-1alpha by other factors, such as c-Myc and phosphorylated Akt (pAkt). This study investigates HIF-1alpha, c-Myc, pAkt, and aerobic glycolysis in low-passage breast cancer cells under the assumption that these represent the in vivo condition better than established lines. Similar to in vivo FdG-PET or primary breast cancers, rates of glycolysis were diverse, being higher in cells expressing both c-Myc and HIF-1alpha and lower in cell lines low or negative in both transcription factors. No correlations were observed between glycolytic rates and pAkt levels. Two of 12 cell lines formed xenografts in mice. Both were positive for HIF-1alpha and phosphorylated c-Myc, and only one was positive for pAkt. Glycolysis was affected by pharmacological regulation of c-Myc and HIF-1alpha. These findings suggest that c-Myc and/or HIF-1alpha activities are both involved in the regulation of glycolysis in breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fluorodeoxyglucose F18/pharmacokinetics , Glucose/pharmacokinetics , Glucose Transporter Type 1/genetics , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/biosynthesis , Mice , Mice, SCID , Phenotype , Phosphorylation , Positron-Emission Tomography/methods , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
8.
Crit Rev Immunol ; 27(3): 221-32, 2007.
Article in English | MEDLINE | ID: mdl-18197818

ABSTRACT

Although the manifestation of inflammatory autodestructive disease is the result of major immunological dysfunction, recent evidence indicates that the immune system attempts to compensate by the production of immunomodulatory autoantibodies. Healthy humans have low levels of naturally occurring autoantibodies directed against the first complementarity-determining region (CDR1) and third framework region (FR3) of their own T-cell receptor (TCR) Vbeta segments, but individuals suffering from rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE) can have highly elevated levels of these autoantibodies. We cloned and characterized human anti-TCR monoclonal autoantibodies (mAAbs) from RA and SLE patients. Because of the cross-reactions between distinct CDR1 segments of human TCR Vbeta and corresponding murine homologs, it was possible to show that human mAAbs blocked the capacity of a murine TH1 cell line (DO11.10) to produce IL-2 in response to antigenic stimulation in vitro. These results support the hypothesis that autoantibodies against TCR Vbeta can shut down TH1-mediated inflammatory autodestructive reactions.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Arthritis, Rheumatoid/immunology , Autoantigens/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Lupus Erythematosus, Systemic/immunology
9.
NMR Biomed ; 18(7): 430-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16206237

ABSTRACT

The hypoxia-inducible transcription factor (HIF-1alpha) plays a central role in tumor development. PX-478 is an experimental anti-cancer drug known to inhibit HIF-1alpha in experimental tumors. The purpose of this study was to identify MRS-visible metabolic biomarkers for PX-478 response prior to phase I/II clinical trials. Single-voxel in vivo localized (1)H spectra were obtained from HT-29 tumor xenografts prior and up to 24 h after treatment with a single dose of PX-478. Profiles of water-soluble and lipophilic metabolites were also examined ex vivo with both (1)H and (31)P spectroscopy for peak identification and to interrogate the underlying biochemistry of the response. The total choline (tCho) resonance was significantly decreased in vivo 12 and 24 h following treatment with PX-478 and this was confirmed with high-resolution (1)H and (31)P MRS. In non-aqueous extracts, significant reductions in cardiolipin, PtdEtn (phosphatidylethanolamine) and PtdI (phosphatidylinositol) were seen in response to PX-478. Although there were trends to a decrease in lactate (and lipid) resonances in vivo and ex vivo, these changes were not significant. This is in contrast to inhibition of in vitro glucose consumption and lactate production by PX-478 in HT-29 cells. The significant and robust change in tCho has identified this as a potential (1)H MRS-visible biomarker for drug response in vivo while high-resolution spectroscopy indicated that GPC, PC, myoI, PE, GPE, CL, PtdEtn and PtdI are potential ex vivo response biomarkers.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mustard Compounds/metabolism , Phenylpropionates/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Choline/chemistry , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Lactic Acid/chemistry , Magnetic Resonance Spectroscopy , Mice , Mice, SCID , Neoplasm Transplantation , Tissue Extracts/chemistry , Transplantation, Heterologous
10.
Neoplasia ; 7(4): 324-30, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15967109

ABSTRACT

Metastatic tumors generally exhibit aerobic glycolysis (the Warburg effect). The advent of [18F]fluorodeoxyglucose positron emission tomography imaging, coupled with recent findings linking hypoxia-inducible factor (HIF-1alpha) overexpression to aggressive cancers, has rekindled an interest in this aspect of tumor metabolism. These studies explore the role of HIF-1alpha in human breast cancer lines and its relationship to glycolytic regulation. Here we demonstrate that, under normal oxygen conditions, nonmetastatic cells consume less glucose and express low HIF-1alpha, whereas metastatic cells constitutively express high glycolysis and HIF-1alpha, suggesting that dysregulation of HIF-1alpha may induce the Warburg effect. This hypothesis was tested by renormalizing HIF-1alpha levels in renal carcinoma cells, leading to inhibition of aerobic glycolysis.


Subject(s)
Gene Expression Regulation, Neoplastic , Glycolysis , Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , DNA Primers/chemistry , Glucose/metabolism , Glucose/pharmacokinetics , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Lactates/metabolism , Neoplasms/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
J Immunol ; 172(7): 4624-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15034081

ABSTRACT

The wide diversity of the T and B Ag receptor repertoires becomes even more extensive postneonatally due to the activity of TdT, which adds nontemplated N nucleotides to Ig and TCR coding ends during V(D)J recombination. In addition, complementarity-determining region 3 sequences formed in the absence of TdT are more uniform due to the use of short sequence homologies between the V, D, and J genes. Thus, the action of TdT produces an adult repertoire that is both different from, and much larger than, the repertoire of the neonate. We have generated TdT-deficient nonobese diabetic (NOD) and MRL-Fas(lpr) mice, and observed a decrease in the incidence of autoimmune disease, including absence of diabetes and decreased pancreatic infiltration in NOD TdT(-/-) mice, and reduced glomerulonephritis and increased life span in MRL-Fas(lpr) TdT(-/-) mice. Using tetramer staining, TdT(-/-) and TdT(+/+) NOD mice showed similar frequencies of the diabetogenic BDC 2.5 CD4(+) T cells. We found no increase in CD4(+)CD25(+) regulatory T cells in NOD TdT(-/-) mice. Thus, TdT deficiency ameliorates the severity of disease in both lupus and diabetes, two very disparate autoimmune diseases that affect different organs, with damage conducted by different effector cell types. The neonatal repertoire appears to be deficient in autoreactive T and/or B cells with high enough affinities to induce end-stage disease. We suggest that the paucity of autoreactive specificities created in the N region-lacking repertoire, and the resultant protection afforded to the newborn, may be the reason that TdT expression is delayed in ontogeny.


Subject(s)
DNA Nucleotidylexotransferase/deficiency , DNA Nucleotidylexotransferase/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , fas Receptor/genetics , Animals , Animals, Newborn , Autoimmune Diseases/genetics , Autoimmune Diseases/mortality , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clone Cells , Crosses, Genetic , DNA Nucleotidylexotransferase/physiology , Diabetes Mellitus, Type 1/mortality , Diabetes Mellitus, Type 1/pathology , Female , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred MRL lpr , Mice, Inbred NOD , Mice, Knockout , Receptors, Interleukin-2/biosynthesis , Survival Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
Immunology ; 105(4): 419-29, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11985662

ABSTRACT

Natural autoantibodies (NAAbs) specific for the T-cell receptor (TCR) are present in all human sera, but individuals with rheumatoid arthritis (RA) generally produce higher titres of immunoglobulin M (IgM) isotype autoantibodies (AAbs) against Vbeta TCR epitopes. To investigate possible correlations between the specificity of such AAbs and their role in immunomodulation, we generated seven B-cell hetero-hybridomas, secreting monoclonal IgM NAAbs, from the synovial tissue and peripheral blood of patients with RA. Here we report three anti-TCR monoclonal autoantibodies (mAAbs)--OR2, OR5 and Syn 2H-11--with the ability to bind subsets of murine T cells, including the ovalbumin-specific DO-11.10 clone. These antibodies did not induce apoptosis in vitro, but prevented interleukin-2 (IL-2) production by antigen-specific T cells. These findings suggest an immunomodulatory function for NAAbs to TCR V-region epitopes and serve as the foundation for testing human anti-TCR mAAbs in animal models with the eventual goal of using them as therapeutic agents in human disease.


Subject(s)
Antibodies, Monoclonal/metabolism , Arthritis, Rheumatoid/immunology , Autoantibodies/metabolism , Interleukin-2/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Autoantibodies/isolation & purification , Cell Line , Clone Cells , Female , Flow Cytometry , Humans , Hybridomas , Mice , Mice, Inbred BALB C , Models, Animal , Protein Binding
13.
FASEB J ; 16(7): 642-52, 2002 May.
Article in English | MEDLINE | ID: mdl-11978728

ABSTRACT

The classical concept of antibody binding is defined as an exclusive and high-affinity interaction with one epitope. The emerging reality about antibody combing sites, however, is that some can bind unrelated determinants. The studies presented here define this quality as epitope recognition promiscuity by analyzing the capacity of monoclonal human autoantibodies to bind sets of overlapping peptides duplicating the complete structures of T cell receptor (TCR) alpha and beta chains and immunoglobulin lambda chain. We assessed the binding of these monoclonal antibodies (mAbs) to a set of homologous peptides corresponding to the CDR1 segments of human Vbeta gene products, a major epitope used in the selection of the antibodies. We present data on the binding characteristics of four human mAbs selected for the ability to bind TCR epitopes. These mAbs are IgM molecules with VH and VL sequences in germline configuration, but have diverse VH CDR3 regions. These studies aim to characterize the property of epitope promiscuity and show that the relationship between the binding site and its epitope is a complex interaction and unpredictable from antigen sequence alone. Our results support the conclusion that epitope recognition promiscuity is a genuine feature of antibody and TCR recognition.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Amino Acid Sequence , Antibody Specificity , Binding Sites , Binding, Competitive , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Epitope Mapping , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...