Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 20(9): 1062-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25129480

ABSTRACT

Technologies are needed to map and image biological barriers in vivo that limit solid tumor delivery and, ultimately, the effectiveness of imaging and therapeutic agents. Here we integrate proteomic and imaging analyses of caveolae at the blood-tumor interface to discover an active transendothelial portal to infiltrate tumors. A post-translationally modified form of annexin A1 (AnnA1) is selectively concentrated in human and rodent tumor caveolae. To follow trafficking, we generated a specific AnnA1 antibody that targets caveolae in the tumor endothelium. Intravital microscopy of caveolae-immunotargeted fluorophores even at low intravenous doses showed rapid and robust pumping across the endothelium to enter mammary, prostate and lung tumors. Within 1 h, the fluorescence signal concentrated throughout tumors to exceed the peak levels in blood. This transvascular pumping required the expression of caveolin 1 and annexin A1. Tumor uptake with other antibodies were >100-fold less. This proteomic imaging strategy reveals a unique target, antibody and caveolae pumping system for solid tumor penetration.


Subject(s)
Caveolae/metabolism , Image Processing, Computer-Assisted/methods , Neoplasms/metabolism , Proteomics , Animals , Humans
2.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L251-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465515

ABSTRACT

Mapping protein expression of endothelial cells (EC) in vivo is fundamental to understanding cellular function and may yield new tissue-selective targets. We have developed a monoclonal antibody, MAb J120, to a protein expressed primarily in rat lung and heart endothelium. The antigen was identified as CD34, a marker of hematopoietic stem cells and global marker of endothelial cells in human and mouse tissues. PCR-based cloning identified two CD34 variant proteins, full length and truncated, both of which are expressed on luminal endothelial cell plasma membranes (P) isolated from lung. Truncated CD34 predominated in heart P, and neither variant was detected in P from kidney or liver. CD34 in lung was readily accessible to (125)I-J120 inoculated intravenously, and immunohistochemistry showed strong CD34 expression in lung EC. Few microvessels stained in heart and kidney, and no CD34 was detected in vessels of other organs or in lymphatics. We present herein the first complete sequence of a rat CD34 variant and show for the first time that the encoded truncated variant is endogenously expressed on EC in vivo. We also demonstrate that CD34 expression in rat EC, unlike mouse and human, is restricted in its distribution enabling quite specific lung targeting in vivo.


Subject(s)
Antigens, CD34/genetics , Antigens, CD34/immunology , Endothelial Cells/physiology , Gene Expression Regulation/immunology , Lung/physiology , Age Factors , Alternative Splicing/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigens, CD34/metabolism , Aorta/cytology , Cells, Cultured , Cloning, Molecular , Endothelial Cells/cytology , Humans , Lung/cytology , Lung/diagnostic imaging , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Rats , Rats, Inbred F344 , Species Specificity , Tomography, Emission-Computed, Single-Photon
3.
J Vasc Res ; 46(4): 311-24, 2009.
Article in English | MEDLINE | ID: mdl-19142011

ABSTRACT

BACKGROUND/AIMS: The vasculature has become an important target in the development of therapies for increasing numbers of human diseases, yet there are few reliable markers available to identify the endothelium in rodent models. We have characterized the expression, subcellular localization and accessibility of the rat pan-endothelial marker podocalyxin (podxl) using a newly developed monoclonal antibody (mAb), G278. METHODS: podxl expression and accessibility to binding by G278 were determined in the rat by a variety of experimental approaches. RESULTS: mAb G278 reliably immunostained blood vessels of all types and of every size in fresh-frozen, fixed-frozen and paraffin-embedded sections of all tissues, but did not stain lymphatic vessels. Western blotting, in vivo imaging and biodistribution analyses demonstrated that the highest levels of endothelial podxl were found in the lung and heart. We also determined that podxl is not enriched in caveolae and that its expression can be modulated in the tumor microenvironment. CONCLUSION: Our study shows that podxl is a better identifier of rat endothelia than are some of the more commonly used markers and that mAb G278 is a robust antibody for use not only in identifying rat blood vessels but also as a tool to elucidate podxl function.


Subject(s)
Endothelium, Vascular/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Sialoglycoproteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Biomarkers/metabolism , Blotting, Western , Brain/blood supply , Caveolae/chemistry , Cell Line, Tumor , Chromatography, Liquid , Coronary Vessels/metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Kidney/blood supply , Liver/blood supply , Lung/blood supply , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Rats , Rats, Inbred F344 , Sialoglycoproteins/immunology , Tandem Mass Spectrometry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
4.
Nat Biotechnol ; 25(3): 327-37, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334358

ABSTRACT

How effectively and quickly endothelial caveolae can transcytose in vivo is unknown, yet critical for understanding their function and potential clinical utility. Here we use quantitative proteomics to identify aminopeptidase P (APP) concentrated in caveolae of lung endothelium. Electron microscopy confirms this and shows that APP antibody targets nanoparticles to caveolae. Dynamic intravital fluorescence microscopy reveals that targeted caveolae operate effectively as pumps, moving antibody within seconds from blood across endothelium into lung tissue, even against a concentration gradient. This active transcytosis requires normal caveolin-1 expression. Whole body gamma-scintigraphic imaging shows rapid, specific delivery into lung well beyond that achieved by standard vascular targeting. This caveolar trafficking in vivo may underscore a key physiological mechanism for selective transvascular exchange and may provide an enhanced delivery system for imaging agents, drugs, gene-therapy vectors and nanomedicines. 'In vivo proteomic imaging' as described here integrates organellar proteomics with multiple imaging techniques to identify an accessible target space that includes the transvascular pumping space of the caveola.


Subject(s)
Aminopeptidases/immunology , Antibodies, Monoclonal/pharmacokinetics , Caveolae/immunology , Endocytosis , Endothelium, Vascular/immunology , Lung/immunology , Video Recording , Animals , Antibodies, Monoclonal/biosynthesis , Caveolae/physiology , Caveolin 1/metabolism , Drug Carriers , Drug Delivery Systems , Endothelium, Vascular/physiology , Lung/blood supply , Lung/physiology , Mice , Nanoparticles , Proteomics/methods , Radionuclide Imaging , Rats , Tomography, Emission-Computed, Single-Photon
5.
Proc Natl Acad Sci U S A ; 103(2): 407-12, 2006 Jan 10.
Article in English | MEDLINE | ID: mdl-16384919

ABSTRACT

The molecular diversity of the luminal endothelial cell surface arising in vivo from local variations in genetic expression and tissue microenvironment may create opportunities for achieving targeted molecular imaging and therapies. Here, we describe a strategy to identify probes and their cognate antigens for targeting vascular endothelia of specific organs in vivo. We differentially screen phage libraries to select organ-targeting antibodies by using luminal endothelial cell plasma membranes isolated directly from tissue and highly enriched in natively expressed proteins exposed to the bloodstream. To obviate liver uptake of intravenously injected phage, we convert the phage-displayed antibodies into scFv-Fc fusion proteins, which then are able to rapidly target select organ(s) in vivo as visualized directly by gamma-scintigraphic whole-body imaging. Mass spectrometry helps identify the antigen targets. This comprehensive strategy provides new promise for harnessing the power of phage display for mapping vascular endothelia natively in tissue and for achieving vascular targeting of specific tissues in vivo.


Subject(s)
Antibodies/analysis , Antibodies/immunology , Endothelial Cells/immunology , Peptide Library , Amino Acid Sequence , Aminopeptidases/immunology , Animals , Antigens/immunology , Endothelial Cells/metabolism , Lung/blood supply , Lung/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Organ Specificity , Rats , Reproducibility of Results
6.
Nat Biotechnol ; 22(8): 985-92, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258593

ABSTRACT

Endothelial cells can function differently in vitro and in vivo; however, the degree of microenvironmental modulation in vivo remains unknown at the molecular level largely because of analytical limitations. We use multidimensional protein identification technology (MudPIT) to identify 450 proteins (with three or more spectra) in luminal endothelial cell plasma membranes isolated from rat lungs and from cultured rat lung microvascular endothelial cells. Forty-one percent of proteins expressed in vivo are not detected in vitro. Statistical analysis measuring reproducibility reveals that seven to ten MudPIT measurements are necessary to achieve > or =95% confidence of analytical completeness with current ion trap equipment. Large-scale mapping of the proteome of vascular endothelial cell surface in vivo, as demonstrated here, is advisable because distinct protein expression is apparently regulated by the tissue microenvironment that cannot yet be duplicated in standard cell culture.


Subject(s)
Endothelial Cells/metabolism , Lung/blood supply , Lung/metabolism , Mass Spectrometry/methods , Membrane Proteins/metabolism , Microcirculation/metabolism , Proteome/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Male , Proteomics/methods , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
7.
Nature ; 429(6992): 629-35, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15190345

ABSTRACT

The molecular complexity of tissues and the inaccessibility of most cells within a tissue limit the discovery of key targets for tissue-specific delivery of therapeutic and imaging agents in vivo. Here, we describe a hypothesis-driven, systems biology approach to identifying a small subset of proteins induced at the tissue-blood interface that are inherently accessible to antibodies injected intravenously. We use subcellular fractionation, subtractive proteomics and bioinformatics to identify endothelial cell surface proteins exhibiting restricted tissue distribution and apparent tissue modulation. Expression profiling and gamma-scintigraphic imaging with antibodies establishes two of these proteins, aminopeptidase-P and annexin A1, as selective in vivo targets for antibodies in lungs and solid tumours, respectively. Radio-immunotherapy to annexin A1 destroys tumours and increases animal survival. This analytical strategy can map tissue- and disease-specific expression of endothelial cell surface proteins to uncover novel accessible targets useful for imaging and therapy.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Profiling , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Proteomics , Radioimmunotherapy/methods , Aminopeptidases/immunology , Aminopeptidases/metabolism , Animals , Annexin A1/immunology , Annexin A1/metabolism , Antibodies/administration & dosage , Antibodies/immunology , Antibodies/therapeutic use , Computational Biology , Drug Delivery Systems/methods , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/radiotherapy , Mass Spectrometry , Membrane Proteins/metabolism , Models, Biological , Organ Specificity , Rats , Survival Rate
8.
Biochem Biophys Res Commun ; 303(3): 733-44, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12670472

ABSTRACT

Monoclonal antibody technology has generated invaluable tools for both the analytical and clinical sciences. However, standard immunization approaches frequently fail to provide monoclonal antibodies with the desired specificity. Subtractive immunization provides a powerful alternative to standard immunization and allows for the production of truly unique antibodies. With the intent of targeting specific epitopes within the proteome, subtractive immunization has been broadly and successfully implemented for the production of monoclonal antibodies otherwise unobtainable by standard immunization. Subtractive immunization utilizes a distinct immune tolerization approach that can substantially enhance the generation of monoclonal antibodies to desired antigens. The approach is based on tolerizing the host animal to immunodominant or otherwise undesired antigen(s) (tolerogen) that may be structurally or functionally related to the antigen of interest. Tolerization of the host animal can be achieved through one of three methods: High Zone, Neonatal, or Drug-induced tolerization. The tolerized animal is then inoculated with the desired antigen (immunogen) and antibodies generated by the subsequent immune response are screened for the desired antigenic reactivity. Over the past 15 years a large number of investigators have used the subtractive approach with cleverly chosen tolerogen-immunogen combinations and successfully generated uniquely reactive antibodies which are often neutralizing or function-blocking. This review will focus on the implementation of subtractive immunization for the production of antibodies otherwise unobtainable by standard immunization.


Subject(s)
Epitopes , Immunization/methods , Proteome , Animals , Animals, Newborn , Antibodies, Monoclonal/biosynthesis , Antibody Specificity , Autoantigens , Cyclophosphamide/pharmacology , Immune Tolerance/drug effects , Immunodominant Epitopes , Immunosuppressive Agents/pharmacology , Mice
9.
Oncogene ; 22(12): 1783-94, 2003 Mar 27.
Article in English | MEDLINE | ID: mdl-12660814

ABSTRACT

We have previously used a subtractive immunization (SI) approach to generate monoclonal antibodies (mAbs) against proteins preferentially expressed by the highly metastatic human epidermoid carcinoma cell line, M(+)HEp3. Here we report the immunopurification, identification and characterization of SIMA135/CDCP1 (subtractive immunization M(+)HEp3 associated 135 kDa protein/CUB domain containing protein 1) using one of these mAbs designated 41-2. Protein expression levels of SIMA135/CDCP1 correlated with the metastatic ability of variant HEp3 cell lines. Protein sequence analysis predicted a cell surface location and type I orientation of SIMA135/CDCP1, which was confirmed directly by immunocytochemistry. Analysis of deglycosylated cell lysates indicated that up to 40 kDa of the apparent molecular weight of SIMA135/CDCP1 is because of N-glycosylation. Western blot analysis using a antiphosphotyrosine antibody demonstrated that SIMA135/CDCP1 from HEp3 cells is tyrosine phosphorylated. Selective inhibitor studies indicated that an Src kinase family member is involved in the tyrosine phosphorylation of the protein. In addition to high expression in M(+)HEp3 cells, the SIMA135/CDCP1 protein is expressed to varying levels in 13 other human tumor cell lines, manifesting only a weak correlation with the reported metastatic ability of these tumor cell lines. The protein is not detected in normal human fibroblasts and endothelial cells. Northern blot analysis indicated that SIMA135/CDCP1 mRNA has a restricted expression pattern in normal human tissues with highest levels of expression in skeletal muscle and colon. Immunohistochemical analysis indicated apical and basal plasma membrane expression of SIMA135/CDCP1 in epithelial cells in normal colon. In colon tumor, SIMA135/CDCP1 expression appeared dysregulated showing extensive cell surface as well as cytoplasmic expression. Consistent with in vitro shedding experiments on HEp3 cells, SIMA135/CDCP1 was also detected within the lumen of normal and cancerous colon crypts, suggesting that protein shedding may occur in vivo. Thus, specific immunodetection followed by proteomic analysis allows for the identification and partial characterization of a heretofore uncharacterized human cell surface antigen.


Subject(s)
Cell Adhesion Molecules/metabolism , Neoplasm Proteins/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antigens, CD , Antigens, Neoplasm , Base Sequence , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , DNA Primers , Humans , Immunohistochemistry , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Phosphorylation , Subtraction Technique , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...