12 results on '"Rafii S"'
Search Results
2. Perforin and serine esterase gene expression in stimulated human T cells. Kinetics, mitogen requirements, and effects of cyclosporin A.
- Author
-
Liu, C C, primary, Rafii, S, additional, Granelli-Piperno, A, additional, Trapani, J A, additional, and Young, J D, additional
- Published
- 1989
- Full Text
- View/download PDF
3. Reversal of emphysema by restoration of pulmonary endothelial cells.
- Author
-
Hisata S, Racanelli AC, Kermani P, Schreiner R, Houghton S, Palikuqi B, Kunar B, Zhou A, McConn K, Capili A, Redmond D, Nolan DJ, Ginsberg M, Ding BS, Martinez FJ, Scandura JM, Cloonan SM, Rafii S, and Choi AMK
- Subjects
- Administration, Intravenous, Animals, Biomarkers metabolism, Disease Models, Animal, Endothelial Cells transplantation, Gene Expression Profiling, Gene Expression Regulation, Glycoproteins metabolism, Humans, Lung blood supply, Lung physiopathology, Mice, Inbred C57BL, Neovascularization, Physiologic, Pancreatic Elastase metabolism, Phenotype, Pulmonary Disease, Chronic Obstructive genetics, Pulmonary Disease, Chronic Obstructive pathology, Pulmonary Disease, Chronic Obstructive physiopathology, Pulmonary Emphysema genetics, Pulmonary Emphysema physiopathology, Severity of Illness Index, Smoking, Transcriptome genetics, Mice, Endothelial Cells pathology, Lung pathology, Pulmonary Emphysema pathology
- Abstract
Chronic obstructive pulmonary disease (COPD) is marked by airway inflammation and airspace enlargement (emphysema) leading to airflow obstruction and eventual respiratory failure. Microvasculature dysfunction is associated with COPD/emphysema. However, it is not known if abnormal endothelium drives COPD/emphysema pathology and/or if correcting endothelial dysfunction has therapeutic potential. Here, we show the centrality of endothelial cells to the pathogenesis of COPD/emphysema in human tissue and using an elastase-induced murine model of emphysema. Airspace disease showed significant endothelial cell loss, and transcriptional profiling suggested an apoptotic, angiogenic, and inflammatory state. This alveolar destruction was rescued by intravenous delivery of healthy lung endothelial cells. Leucine-rich α-2-glycoprotein-1 (LRG1) was a driver of emphysema, and deletion of Lrg1 from endothelial cells rescued vascular rarefaction and alveolar regression. Hence, targeting endothelial cell biology through regenerative methods and/or inhibition of the LRG1 pathway may represent strategies of immense potential for the treatment of COPD/emphysema., Competing Interests: Disclosures: D.J. Nolan reported personal fees from Angiocrine Bioscience outside the submitted work; in addition, D.J. Nolan had a patent number 8,465,732 issued (Angiocrine Bioscience) and a patent number 9,944,897 issued; and is an employee and equity holder of Angiocrine Bioscience. M. Ginsberg reported personal fees from Angiocrine Bioscience outside the submitted work; in addition, M. Ginsberg had a patent to 8,465,732 issued and a patent to 9,944,897 issued. In addition, M. Ginsberg is a current employee and equity holder of Angiocrine Bioscience. F.J. Martinez reported non-financial support from ProterrixBio, Nitto, Zambon; "other" from Afferent/Merck, Biogen, Veracyte, Prometic, Bridge Biotherapeutics, and Abbvie; grants from Gilead; and personal fees from AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Chiesi, Sunovion, Patara/Respivant, Bayer, Promedior/Roche, Teva, Col Behring, DevPro, IQVIA, Sanofi/Regeneron, United Therapeutics, and Novartis outside the submitted work. S.M. Cloonan reported grants from National Institute of Health, National Heart, Blood and Lung Institute (NHLBI), and Science Foundation Ireland (SFI), and personal fees from Pharmacosmos outside the submitted work; in addition, S.M. Cloonan had a patent number 10,905,682 issued. S. Rafii reported non-financial support from Angiocrine Bioscience during the conduct of the study; non-financial support from Angiocrine Bioscience outside the submitted work; and had a patent to E4ORF1 Endothelial cell infusion for organ repair licensed (Angiocrine Bioscience). A.M.K. Choi is a cofounder and equity stock holder for Proterris, which develops therapeutic uses for carbon monoxide. A.M.K. Choi has a use patent on CO. Additionally, A.M.K. Choi has a patent in COPD. No other disclosures were reported., (© 2021 Hisata et al.)
- Published
- 2021
- Full Text
- View/download PDF
4. Single-cell profiling reveals an endothelium-mediated immunomodulatory pathway in the eye choroid.
- Author
-
Lehmann GL, Hanke-Gogokhia C, Hu Y, Bareja R, Salfati Z, Ginsberg M, Nolan DJ, Mendez-Huergo SP, Dalotto-Moreno T, Wojcinski A, Ochoa F, Zeng S, Cerliani JP, Panagis L, Zager PJ, Mullins RF, Ogura S, Lutty GA, Bang J, Zippin JH, Romano C, Rabinovich GA, Elemento O, Joyner AL, Rafii S, Rodriguez-Boulan E, and Benedicto I
- Subjects
- Animals, Cell Proliferation, Endothelial Cells metabolism, Gene Expression Regulation, Hedgehog Proteins metabolism, Inflammation genetics, Mast Cells metabolism, Melanocytes metabolism, Melanocytes pathology, Mice, Inbred C57BL, Organ Specificity, Retinal Pigment Epithelium metabolism, Signal Transduction, Transcription, Genetic, Zinc Finger Protein GLI1 metabolism, Choroid immunology, Choroid pathology, Endothelium immunology, Immunomodulation, Single-Cell Analysis
- Abstract
The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders., Competing Interests: Disclosures: Dr. Nolan reported personal fees from Angiocrine Bioscience during the conduct of the study; in addition, Dr. Nolan had a patent number 9,944,897 issued "Angiocrine Bioscience." Dr. Panagis reported personal fees from Regeneron outside the submitted work. Dr. Zippin reported grants from Pfizer and personal fees from Hoth outside the submitted work. Dr. Romano reported, "I am an employee of Regeneron Pharmaceuticals, however there are no products or services of this company related to the work presented in this manuscript." Dr. Elemento reported "other" from Volastra Therapeutics, "other" from One Three Biotech, "other" from Freenome, and "other" from Owkin outside the submitted work. Dr. Rafii reported non-financial support from Angiocrine BioScience during the conduct of the study. No other disclosures were reported., (© 2020 Lehmann et al.)
- Published
- 2020
- Full Text
- View/download PDF
5. An activated form of ADAM10 is tumor selective and regulates cancer stem-like cells and tumor growth.
- Author
-
Atapattu L, Saha N, Chheang C, Eissman MF, Xu K, Vail ME, Hii L, Llerena C, Liu Z, Horvay K, Abud HE, Kusebauch U, Moritz RL, Ding BS, Cao Z, Rafii S, Ernst M, Scott AM, Nikolov DB, Lackmann M, and Janes PW
- Subjects
- ADAM10 Protein antagonists & inhibitors, ADAM10 Protein chemistry, ADAM17 Protein physiology, Amino Acid Motifs, Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal immunology, Humans, Male, Mice, Mice, Inbred BALB C, Receptors, Notch physiology, ADAM10 Protein physiology, Neoplasms, Experimental pathology, Neoplastic Stem Cells pathology
- Abstract
The transmembrane metalloprotease ADAM10 sheds a range of cell surface proteins, including ligands and receptors of the Notch, Eph, and erbB families, thereby activating signaling pathways critical for tumor initiation and maintenance. ADAM10 is thus a promising therapeutic target. Although widely expressed, its activity is normally tightly regulated. We now report prevalence of an active form of ADAM10 in tumors compared with normal tissues, in mouse models and humans, identified by our conformation-specific antibody mAb 8C7. Structure/function experiments indicate mAb 8C7 binds an active conformation dependent on disulfide isomerization and oxidative conditions, common in tumors. Moreover, this active ADAM10 form marks cancer stem-like cells with active Notch signaling, known to mediate chemoresistance. Importantly, specific targeting of active ADAM10 with 8C7 inhibits Notch activity and tumor growth in mouse models, particularly regrowth after chemotherapy. Our results indicate targeted inhibition of active ADAM10 as a potential therapy for ADAM10-dependent tumor development and drug resistance., (© 2016 Atapattu et al.)
- Published
- 2016
- Full Text
- View/download PDF
6. TGFβ restores hematopoietic homeostasis after myelosuppressive chemotherapy.
- Author
-
Brenet F, Kermani P, Spektor R, Rafii S, and Scandura JM
- Subjects
- Animals, Cyclin-Dependent Kinase Inhibitor p57 analysis, Hematopoietic Stem Cells physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction, Transforming Growth Factor beta antagonists & inhibitors, Antineoplastic Agents toxicity, Bone Marrow drug effects, Hematopoiesis drug effects, Homeostasis, Transforming Growth Factor beta physiology
- Abstract
Myelosuppression is a life-threatening complication of antineoplastic therapy, but treatment is restricted to a few cytokines with unilineage hematopoietic activity. Although hematopoietic stem cells (HSCs) are predominantly quiescent during homeostasis, they are rapidly recruited into cell cycle by stresses, including myelosuppressive chemotherapy. Factors that induce HSCs to proliferate during stress have been characterized, but it is not known how HSC quiescence is then reestablished. In this study, we show that TGFβ signaling is transiently activated in hematopoietic stem and progenitor cells (HSPCs) during hematopoietic regeneration. Blockade of TGFβ signaling after chemotherapy accelerates hematopoietic reconstitution and delays the return of cycling HSCs to quiescence. In contrast, TGFβ blockade during homeostasis fails to induce cycling of HSPCs. We identified the cyclin-dependent kinase inhibitor Cdkn1c (p57) as a key downstream mediator of TGFβ during regeneration because the recovery of chimeric mice, incapable of expressing p57 in HSPCs, phenocopies blockade of TGFβ signaling after chemotherapy. This study demonstrates that context-dependent activation of TGFβ signaling is central to an unrecognized counterregulatory mechanism that promotes homeostasis once hematopoiesis has sufficiently recovered from myelosuppressive chemotherapy. These results open the door to new, potentially superior, approaches to promote multilineage hematopoietic recovery by blocking the TGFβ signaling that dampens regeneration.
- Published
- 2013
- Full Text
- View/download PDF
7. S1P and the birth of platelets.
- Author
-
Hla T, Galvani S, Rafii S, and Nachman R
- Subjects
- Blood Platelets metabolism, Chemokine CXCL12 metabolism, Extracellular Fluid metabolism, Lysophospholipids blood, Lysophospholipids metabolism, Receptors, CXCR4 metabolism, Sphingosine blood, Sphingosine metabolism, Sphingosine physiology, Blood Platelets physiology, Cell Movement physiology, Lysophospholipids physiology, Models, Biological, Receptors, Lysosphingolipid metabolism, Sphingosine analogs & derivatives, Thrombopoiesis physiology
- Abstract
Recent work has highlighted the multitude of biological functions of sphingosine 1-phosphate (S1P), which include roles in hematopoietic cell trafficking, organization of immune organs, vascular development, and neuroinflammation. Indeed, a functional antagonist of S1P(1) receptor, FTY720/Gilenya, has entered the clinic as a novel therapeutic for multiple sclerosis. In this issue of the JEM, Zhang et al. highlight yet another function of this lipid mediator: thrombopoiesis. The S1P(1) receptor is required for the growth of proplatelet strings in the bloodstream and the shedding of platelets into the circulation. Notably, the sharp gradient of S1P between blood and the interstitial fluids seems to be essential to ensure the production of platelets, and S1P appears to cooperate with the CXCL12-CXCR4 axis. Pharmacologic modulation of the S1P(1) receptor altered circulating platelet numbers acutely, suggesting a potential therapeutic strategy for controlling thrombocytopenic states. However, the S1P(4) receptor may also regulate thrombopoiesis during stress-induced accelerated platelet production. This work reveals a novel physiological action of the S1P/S1P(1) duet that could potentially be harnessed for clinical translation.
- Published
- 2012
- Full Text
- View/download PDF
8. Low-dose irradiation promotes tissue revascularization through VEGF release from mast cells and MMP-9-mediated progenitor cell mobilization.
- Author
-
Heissig B, Rafii S, Akiyama H, Ohki Y, Sato Y, Rafael T, Zhu Z, Hicklin DJ, Okumura K, Ogawa H, Werb Z, and Hattori K
- Subjects
- Animals, Bone Marrow radiation effects, Cell Proliferation radiation effects, Extremities blood supply, Hematopoietic Stem Cells enzymology, Ischemia radiotherapy, Mast Cells enzymology, Matrix Metalloproteinase 9 deficiency, Matrix Metalloproteinase 9 genetics, Matrix Metalloproteinase 9 radiation effects, Mice, Gamma Rays, Hematopoietic Stem Cells radiation effects, Mast Cells metabolism, Mast Cells radiation effects, Matrix Metalloproteinase 9 physiology, Neovascularization, Physiologic radiation effects, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A radiation effects
- Abstract
Mast cells accumulate in tissues undergoing angiogenesis during tumor growth, wound healing, and tissue repair. Mast cells can secrete angiogenic factors such as vascular endothelial growth factor (VEGF). Ionizing irradiation has also been shown to have angiogenic potential in malignant and nonmalignant diseases. We observed that low-dose irradiation fosters mast cell-dependent vascular regeneration in a limb ischemia model. Irradiation promoted VEGF production by mast cells in a matrix metalloproteinase-9 (MMP-9)-dependent manner. Irradiation, through MMP-9 up-regulated by VEGF in stromal and endothelial cells, induced the release of Kit-ligand (KitL). Irradiation-induced VEGF promoted migration of mast cells from the bone marrow to the ischemic site. Irradiation-mediated release of KitL and VEGF was impaired in MMP-9-deficient mice, resulting in a reduced number of tissue mast cells and delayed vessel formation in the ischemic limb. Irradiation-induced vasculogenesis was abrogated in mice deficient in mast cells (steel mutant, Sl/Sl(d) mice) and in mice in which the VEGF pathway was blocked. Irradiation did not induce progenitor mobilization in Sl/Sl(d) mice. We conclude that increased recruitment and activation of mast cells following irradiation alters the ischemic microenvironment and promotes vascular regeneration in an ischemia model. These data show a novel mechanism of neovascularization and suggest that low-dose irradiation may be used for therapeutic angiogenesis to augment vasculogenesis in ischemic tissues.
- Published
- 2005
- Full Text
- View/download PDF
9. CD44 is a major E-selectin ligand on human hematopoietic progenitor cells.
- Author
-
Dimitroff CJ, Lee JY, Rafii S, Fuhlbrigge RC, and Sackstein R
- Subjects
- Animals, Blotting, Western methods, CHO Cells, Cricetinae, E-Selectin genetics, Electrophoresis, Polyacrylamide Gel methods, HL-60 Cells, Hematopoietic Stem Cells cytology, Humans, Hyaluronan Receptors genetics, K562 Cells, Ligands, Membrane Glycoproteins metabolism, P-Selectin metabolism, Sodium Dodecyl Sulfate, E-Selectin metabolism, Hematopoietic Stem Cells metabolism, Hyaluronan Receptors metabolism
- Abstract
E-selectin plays a critical role in mediating tissue-specific homing of T cells into skin, and of primitive hematopoietic progenitor cells (HPCs) into bone marrow (BM). Though it is known that a glycoform of PSGL-1 (CLA) functions as the principal E-selectin ligand on human T lymphocytes, the E-selectin ligand(s) of human HPCs has not been identified. We used a shear-based adherence assay to analyze and define the E-selectin ligand activity of membrane proteins from human HPCs. Our data show that PSGL-1 expressed on human HPCs is an E-selectin ligand, and that HPCs also express a previously unrecognized E-selectin ligand, CD44. The E-selectin ligand activity of CD44 is conferred by the elaboration of sialylated, fucosylated binding determinants on N-glycans. This glycoform of CD44 is expressed on primitive CD34+ human HPCs, but not on more mature hematopoietic cells. Under physiologic flow conditions, this molecule mediates E-selectin-dependent rolling interactions over a wider shear range than that of PSGL-1, and promotes human HPC rolling interactions on E-selectin expressed on human BM endothelial cells. These findings offer new insights into the structural biology and physiology of CD44, and into the molecular basis of E-selectin-dependent adhesive interactions that direct homing of human HPC to BM.
- Published
- 2001
- Full Text
- View/download PDF
10. Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells.
- Author
-
Hattori K, Dias S, Heissig B, Hackett NR, Lyden D, Tateno M, Hicklin DJ, Zhu Z, Witte L, Crystal RG, Moore MA, and Rafii S
- Subjects
- Adenoviridae, Angiopoietin-1, Animals, Bone Marrow Cells, Endothelial Growth Factors administration & dosage, Endothelial Growth Factors blood, Endothelial Growth Factors metabolism, Female, Genetic Vectors, Hematopoietic Stem Cell Mobilization, Leukocytes physiology, Lymphokines administration & dosage, Lymphokines blood, Lymphokines metabolism, Male, Membrane Glycoproteins administration & dosage, Membrane Glycoproteins blood, Membrane Glycoproteins metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, SCID, Neoplasm Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Spleen cytology, Time Factors, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors physiology, Hematopoiesis physiology, Hematopoietic Stem Cells physiology, Lymphokines physiology, Membrane Glycoproteins physiology, Proto-Oncogene Proteins, Signal Transduction physiology
- Abstract
Tyrosine kinase receptors for angiogenic factors vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) are expressed not only by endothelial cells but also by subsets of hematopoietic stem cells (HSCs). To further define their role in the regulation of postnatal hematopoiesis and vasculogenesis, VEGF and Ang-1 plasma levels were elevated by injecting recombinant protein or adenoviral vectors expressing soluble VEGF(165), matrix-bound VEGF(189), or Ang-1 into mice. VEGF(165), but not VEGF(189), induced a rapid mobilization of HSCs and VEGF receptor (VEGFR)2(+) circulating endothelial precursor cells (CEPs). In contrast, Ang-1 induced delayed mobilization of CEPs and HSCs. Combined sustained elevation of Ang-1 and VEGF(165) was associated with an induction of hematopoiesis and increased marrow cellularity followed by proliferation of capillaries and expansion of sinusoidal space. Concomitant to this vascular remodeling, there was a transient depletion of hematopoietic activity in the marrow, which was compensated by an increase in mobilization and recruitment of HSCs and CEPs to the spleen resulting in splenomegaly. Neutralizing monoclonal antibody to VEGFR2 completely inhibited VEGF(165), but not Ang-1-induced mobilization and splenomegaly. These data suggest that temporal and regional activation of VEGF/VEGFR2 and Ang-1/Tie-2 signaling pathways are critical for mobilization and recruitment of HSCs and CEPs and may play a role in the physiology of postnatal angiogenesis and hematopoiesis.
- Published
- 2001
- Full Text
- View/download PDF
11. Transendothelial migration of megakaryocytes in response to stromal cell-derived factor 1 (SDF-1) enhances platelet formation.
- Author
-
Hamada T, Möhle R, Hesselgesser J, Hoxie J, Nachman RL, Moore MA, and Rafii S
- Subjects
- Cell Line, Chemokine CXCL12, Humans, Megakaryocytes drug effects, Polyploidy, Receptors, CXCR4 biosynthesis, Blood Platelets physiology, Bone Marrow physiology, Chemokines, CXC physiology, Chemotaxis physiology, Endothelium, Vascular physiology, Megakaryocytes physiology
- Abstract
Although thrombopoietin has been shown to promote megakaryocyte (MK) proliferation and maturation, the exact mechanism and site of platelet formation are not well defined. Studies have shown that MKs may transmigrate through bone marrow endothelial cells (BMEC), and release platelets within the sinusoidal space or lung capillaries. In search for chemotactic factor(s) that may mediate transmigration of MKs, we have discovered that mature polyploid MKs express the G protein-coupled chemokine receptor CXCR4 (Fusin, LESTR). Therefore, we explored the possibility that stromal cell-derived factor 1 (SDF-1), the ligand for CXCR4, may also induce transendothelial migration of mature MKs. SDF-1, but not other CXC or CC chemokines, was able to mediate MK migration (ED50 = 125 pmol/liter). The MK chemotaxis induced by SDF-1 was inhibited by the CXCR4-specific mAb (12G5) and by pertussis toxin, demonstrating that signaling via the G protein-coupled receptor CXCR4 was necessary for migration. SDF-1 also induced MKs to migrate through confluent monolayers of BMEC by increasing the affinity of MKs for BMEC. Activation of BMEC with interleukin 1beta resulted in a threefold increase in the migration of MKs in response to SDF-1. Neutralizing mAb to the endothelial-specific adhesion molecule E-selectin blocked the migration of MKs by 50%, suggesting that cellular interaction of MKs with BMEC is critical for the migration of MKs. Light microscopy and ploidy determination of transmigrated MKs demonstrated predominance of polyploid MKs. Virtually all platelets generated in the lower chamber also expressed CXCR4. Platelets formed in the lower chamber were functional and expressed P-selectin (CD62P) in response to thrombin stimulation. Electron microscopy of the cells that transmigrated through the BMEC monolayers in response to SDF-1 demonstrated the presence of intact polyploid MKs as well as MKs in the process of platelet formation. These results suggest that SDF-1 is a potent chemotactic factor for mature MKs. Expression of CXCR4 may be the critical cellular signal for transmigration of MKs and platelet formation.
- Published
- 1998
- Full Text
- View/download PDF
12. Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model antigen induce protective and therapeutic antitumor immunity.
- Author
-
Song W, Kong HL, Carpenter H, Torii H, Granstein R, Rafii S, Moore MA, and Crystal RG
- Subjects
- Adenocarcinoma prevention & control, Adenocarcinoma therapy, Adenoviridae immunology, Animals, Bone Marrow Cells immunology, Bone Marrow Cells virology, Bone Marrow Transplantation, Cell Line, Colonic Neoplasms prevention & control, Colonic Neoplasms therapy, Dendritic Cells virology, Gene Transfer Techniques, Genetic Vectors immunology, Humans, Lung Neoplasms immunology, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Lymphocyte Activation genetics, Male, Mice, Mice, Inbred BALB C, Models, Immunological, Neoplasm Transplantation, T-Lymphocytes, Cytotoxic immunology, Adenocarcinoma immunology, Adenoviridae genetics, Colonic Neoplasms immunology, DNA, Complementary immunology, Dendritic Cells immunology, Dendritic Cells transplantation, Immunotherapy, Adoptive methods, beta-Galactosidase immunology
- Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in the initiation of antitumor immune responses. In this study, we show that genetic modifications of a murine epidermis-derived DC line and primary bone marrow-derived DCs to express a model antigen beta-galactosidase (betagal) can be achieved through the use of a replication-deficient, recombinant adenovirus vector, and that the modified DCs are capable of eliciting antigen-specific, MHC-restricted CTL responses. Importantly, using a murine metastatic lung tumor model with syngeneic colon carcinoma cells expressing betagal, we show that immunization of mice with the genetically modified DC line or bone marrow DCs confers potent protection against a lethal tumor challenge, as well as suppression of preestablished tumors, resulting in a significant survival advantage. We conclude that genetic modification of DCs to express antigens that are also expressed in tumors can lead to antigen-specific, antitumor killer cells, with a concomitant resistance to tumor challenge and a decrease in the size of existing tumors.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.