Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes

Background: In the present study, we demonstrate, in rigorous fashion, that human monocyte-derived immature dendritic cells (DCs) can efficiently cross-present tumor-associated antigens when co-cultured with a mixture of human melanoma cells rendered apoptotic/necrotic by γ irradiation (Apo-Nec cell...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: von Euw, E.M., Barrio, M.M., Furman, D., Bianchini, M., Levy, E.M., Yee, C., Li, Y., Wainstok, R., Mordoh, J.
Formato: Artículo publishedVersion
Lenguaje:Inglés
Publicado: 2007
Materias:
Acceso en línea:http://hdl.handle.net/20.500.12110/paper_14795876_v5_n_p_vonEuw
Aporte de:
id paperaa:paper_14795876_v5_n_p_vonEuw
record_format dspace
institution Universidad de Buenos Aires
institution_str I-28
repository_str R-134
collection Biblioteca Digital - Facultad de Ciencias Exactas y Naturales (UBA)
language Inglés
orig_language_str_mv eng
topic B7 antigen
cancer vaccine
CD40 antigen
CD83 antigen
CD86 antigen
chemokine
chemokine receptor CCR7
dendritic cell vaccine
dextran
fluorescein isothiocyanate
gamma interferon
glycoprotein gp 100
HLA A antigen
HLA antigen
HLA antigen class 1
HLA antigen class 2
HLA antigen class 3
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
tumor antigen
unclassified drug
CCR7 protein, human
chemokine receptor CCR7
epitope
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
melanocyte protein Pmel 17
membrane protein
MLANA protein, human
SILV protein, human
tumor antigen
tumor protein
antigen expression
antigen specificity
apoptosis
article
CD8+ T lymphocyte
cell cloning
cell death
cell kinetics
cell maturation
cell migration
cell surface
cell vacuole
coculture
controlled study
cross presentation
cytokine release
cytotoxic T lymphocyte
dendritic cell
electron microscopy
fluorescence activated cell sorting
gamma irradiation
human
human cell
in vitro study
melanoma cell
monocyte
phagocytosis
upregulation
vaccination
apoptosis
cell differentiation
cell motion
cross presentation
cytology
drug effect
gamma radiation
immunology
melanoma
monocyte
necrosis
pathology
radiation exposure
time
tumor cell line
ultrastructure
Antigens, Neoplasm
Apoptosis
CD8-Positive T-Lymphocytes
Cell Differentiation
Cell Line, Tumor
Cell Movement
Chemokine CCL19
Coculture Techniques
Cross-Priming
Dendritic Cells
Epitopes
Gamma Rays
gp100 Melanoma Antigen
Humans
Interleukin-10
Interleukin-12
MART-1 Antigen
Melanoma
Membrane Glycoproteins
Monocytes
Necrosis
Neoplasm Proteins
Phagocytosis
Receptors, CCR7
Time Factors
spellingShingle B7 antigen
cancer vaccine
CD40 antigen
CD83 antigen
CD86 antigen
chemokine
chemokine receptor CCR7
dendritic cell vaccine
dextran
fluorescein isothiocyanate
gamma interferon
glycoprotein gp 100
HLA A antigen
HLA antigen
HLA antigen class 1
HLA antigen class 2
HLA antigen class 3
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
tumor antigen
unclassified drug
CCR7 protein, human
chemokine receptor CCR7
epitope
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
melanocyte protein Pmel 17
membrane protein
MLANA protein, human
SILV protein, human
tumor antigen
tumor protein
antigen expression
antigen specificity
apoptosis
article
CD8+ T lymphocyte
cell cloning
cell death
cell kinetics
cell maturation
cell migration
cell surface
cell vacuole
coculture
controlled study
cross presentation
cytokine release
cytotoxic T lymphocyte
dendritic cell
electron microscopy
fluorescence activated cell sorting
gamma irradiation
human
human cell
in vitro study
melanoma cell
monocyte
phagocytosis
upregulation
vaccination
apoptosis
cell differentiation
cell motion
cross presentation
cytology
drug effect
gamma radiation
immunology
melanoma
monocyte
necrosis
pathology
radiation exposure
time
tumor cell line
ultrastructure
Antigens, Neoplasm
Apoptosis
CD8-Positive T-Lymphocytes
Cell Differentiation
Cell Line, Tumor
Cell Movement
Chemokine CCL19
Coculture Techniques
Cross-Priming
Dendritic Cells
Epitopes
Gamma Rays
gp100 Melanoma Antigen
Humans
Interleukin-10
Interleukin-12
MART-1 Antigen
Melanoma
Membrane Glycoproteins
Monocytes
Necrosis
Neoplasm Proteins
Phagocytosis
Receptors, CCR7
Time Factors
von Euw, E.M.
Barrio, M.M.
Furman, D.
Bianchini, M.
Levy, E.M.
Yee, C.
Li, Y.
Wainstok, R.
Mordoh, J.
Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
topic_facet B7 antigen
cancer vaccine
CD40 antigen
CD83 antigen
CD86 antigen
chemokine
chemokine receptor CCR7
dendritic cell vaccine
dextran
fluorescein isothiocyanate
gamma interferon
glycoprotein gp 100
HLA A antigen
HLA antigen
HLA antigen class 1
HLA antigen class 2
HLA antigen class 3
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
tumor antigen
unclassified drug
CCR7 protein, human
chemokine receptor CCR7
epitope
interleukin 10
interleukin 12
macrophage inflammatory protein 3beta
melan A
melanocyte protein Pmel 17
membrane protein
MLANA protein, human
SILV protein, human
tumor antigen
tumor protein
antigen expression
antigen specificity
apoptosis
article
CD8+ T lymphocyte
cell cloning
cell death
cell kinetics
cell maturation
cell migration
cell surface
cell vacuole
coculture
controlled study
cross presentation
cytokine release
cytotoxic T lymphocyte
dendritic cell
electron microscopy
fluorescence activated cell sorting
gamma irradiation
human
human cell
in vitro study
melanoma cell
monocyte
phagocytosis
upregulation
vaccination
apoptosis
cell differentiation
cell motion
cross presentation
cytology
drug effect
gamma radiation
immunology
melanoma
monocyte
necrosis
pathology
radiation exposure
time
tumor cell line
ultrastructure
Antigens, Neoplasm
Apoptosis
CD8-Positive T-Lymphocytes
Cell Differentiation
Cell Line, Tumor
Cell Movement
Chemokine CCL19
Coculture Techniques
Cross-Priming
Dendritic Cells
Epitopes
Gamma Rays
gp100 Melanoma Antigen
Humans
Interleukin-10
Interleukin-12
MART-1 Antigen
Melanoma
Membrane Glycoproteins
Monocytes
Necrosis
Neoplasm Proteins
Phagocytosis
Receptors, CCR7
Time Factors
description Background: In the present study, we demonstrate, in rigorous fashion, that human monocyte-derived immature dendritic cells (DCs) can efficiently cross-present tumor-associated antigens when co-cultured with a mixture of human melanoma cells rendered apoptotic/necrotic by γ irradiation (Apo-Nec cells). Methods: We evaluated the phagocytosis of Apo-Nec cells by FACS after PKH26 and PKH67 staining of DCs and Apo-Nec cells at different times of coculture. The kinetics of the process was also followed by electron microscopy. DCs maturation was also studied monitoring the expression of specific markers, migration towards specific chemokines and the ability to cross-present in vitro the native melanoma-associated Ags MelanA/MART-1 and gp100. Results: Apo-Nec cells were efficiently phagocytosed by immature DCs (iDC) (55 ± 10.5%) at 12 hs of coculture. By 12-24 hs we observed digested Apo-Nec cells inside DCs and large empty vacuoles as part of the cellular processing. Loading with Apo-Nec cells induced DCs maturation to levels achieved using LPS treatment, as measured by: i) the decrease in FITC - Dextran uptake (iDC: 81 ± 5%; DC/Apo-Nec 33 ± 12%); ii) the cell surface up-regulation of CD80, CD86, CD83, CCR7, CD40, HLA-I and HLA-II and iii) an increased in vitro migration towards MIP-3β. DC/Apo-Nec isolated from HLA-A*0201 donors were able to induce >600 pg/ml IFN-γ secretion of CTL clones specific for MelanA/MART-1 and gp100 Ags after 6 hs and up to 48 hs of coculture, demonstrating efficient cross-presentation of the native Ags. Intracellular IL-12 was detected in DC/Apo-Nec 24 hs post-coculture while IL-10 did not change. Conclusion: We conclude thatthe use of a mixture of four apoptotic/ necrotic melanoma cell lines is a suitable source of native melanoma Ags that provides maturation signals for DCs, increases migration to MIP-3β and allows Ag cross-presentation. This strategy could be exploited for vaccination of melanoma patients. © 2007 von Euw et al; licensee BioMed Central Ltd.
format Artículo
Artículo
publishedVersion
author von Euw, E.M.
Barrio, M.M.
Furman, D.
Bianchini, M.
Levy, E.M.
Yee, C.
Li, Y.
Wainstok, R.
Mordoh, J.
author_facet von Euw, E.M.
Barrio, M.M.
Furman, D.
Bianchini, M.
Levy, E.M.
Yee, C.
Li, Y.
Wainstok, R.
Mordoh, J.
author_sort von Euw, E.M.
title Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
title_short Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
title_full Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
title_fullStr Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
title_full_unstemmed Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes
title_sort monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and mart-1 antigens to specific cd8+ t lymphocytes
publishDate 2007
url http://hdl.handle.net/20.500.12110/paper_14795876_v5_n_p_vonEuw
work_keys_str_mv AT voneuwem monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT barriomm monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT furmand monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT bianchinim monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT levyem monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT yeec monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT liy monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT wainstokr monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
AT mordohj monocytederiveddendriticcellsloadedwithamixtureofapoptoticnecroticmelanomacellsefficientlycrosspresentgp100andmart1antigenstospecificcd8tlymphocytes
_version_ 1769810045029056512
spelling paperaa:paper_14795876_v5_n_p_vonEuw2023-06-12T16:50:29Z Monocyte-derived dendritic cells loaded with a mixture of apoptotic/ necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8+ T lymphocytes J. Transl. Med. 2007;5 von Euw, E.M. Barrio, M.M. Furman, D. Bianchini, M. Levy, E.M. Yee, C. Li, Y. Wainstok, R. Mordoh, J. B7 antigen cancer vaccine CD40 antigen CD83 antigen CD86 antigen chemokine chemokine receptor CCR7 dendritic cell vaccine dextran fluorescein isothiocyanate gamma interferon glycoprotein gp 100 HLA A antigen HLA antigen HLA antigen class 1 HLA antigen class 2 HLA antigen class 3 interleukin 10 interleukin 12 macrophage inflammatory protein 3beta melan A tumor antigen unclassified drug CCR7 protein, human chemokine receptor CCR7 epitope interleukin 10 interleukin 12 macrophage inflammatory protein 3beta melan A melanocyte protein Pmel 17 membrane protein MLANA protein, human SILV protein, human tumor antigen tumor protein antigen expression antigen specificity apoptosis article CD8+ T lymphocyte cell cloning cell death cell kinetics cell maturation cell migration cell surface cell vacuole coculture controlled study cross presentation cytokine release cytotoxic T lymphocyte dendritic cell electron microscopy fluorescence activated cell sorting gamma irradiation human human cell in vitro study melanoma cell monocyte phagocytosis upregulation vaccination apoptosis cell differentiation cell motion cross presentation cytology drug effect gamma radiation immunology melanoma monocyte necrosis pathology radiation exposure time tumor cell line ultrastructure Antigens, Neoplasm Apoptosis CD8-Positive T-Lymphocytes Cell Differentiation Cell Line, Tumor Cell Movement Chemokine CCL19 Coculture Techniques Cross-Priming Dendritic Cells Epitopes Gamma Rays gp100 Melanoma Antigen Humans Interleukin-10 Interleukin-12 MART-1 Antigen Melanoma Membrane Glycoproteins Monocytes Necrosis Neoplasm Proteins Phagocytosis Receptors, CCR7 Time Factors Background: In the present study, we demonstrate, in rigorous fashion, that human monocyte-derived immature dendritic cells (DCs) can efficiently cross-present tumor-associated antigens when co-cultured with a mixture of human melanoma cells rendered apoptotic/necrotic by γ irradiation (Apo-Nec cells). Methods: We evaluated the phagocytosis of Apo-Nec cells by FACS after PKH26 and PKH67 staining of DCs and Apo-Nec cells at different times of coculture. The kinetics of the process was also followed by electron microscopy. DCs maturation was also studied monitoring the expression of specific markers, migration towards specific chemokines and the ability to cross-present in vitro the native melanoma-associated Ags MelanA/MART-1 and gp100. Results: Apo-Nec cells were efficiently phagocytosed by immature DCs (iDC) (55 ± 10.5%) at 12 hs of coculture. By 12-24 hs we observed digested Apo-Nec cells inside DCs and large empty vacuoles as part of the cellular processing. Loading with Apo-Nec cells induced DCs maturation to levels achieved using LPS treatment, as measured by: i) the decrease in FITC - Dextran uptake (iDC: 81 ± 5%; DC/Apo-Nec 33 ± 12%); ii) the cell surface up-regulation of CD80, CD86, CD83, CCR7, CD40, HLA-I and HLA-II and iii) an increased in vitro migration towards MIP-3β. DC/Apo-Nec isolated from HLA-A*0201 donors were able to induce >600 pg/ml IFN-γ secretion of CTL clones specific for MelanA/MART-1 and gp100 Ags after 6 hs and up to 48 hs of coculture, demonstrating efficient cross-presentation of the native Ags. Intracellular IL-12 was detected in DC/Apo-Nec 24 hs post-coculture while IL-10 did not change. Conclusion: We conclude thatthe use of a mixture of four apoptotic/ necrotic melanoma cell lines is a suitable source of native melanoma Ags that provides maturation signals for DCs, increases migration to MIP-3β and allows Ag cross-presentation. This strategy could be exploited for vaccination of melanoma patients. © 2007 von Euw et al; licensee BioMed Central Ltd. Fil:von Euw, E.M. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentina. Fil:Barrio, M.M. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentina. Fil:Levy, E.M. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentina. 2007 info:eu-repo/semantics/article info:ar-repo/semantics/artículo info:eu-repo/semantics/publishedVersion application/pdf eng info:eu-repo/semantics/openAccess http://creativecommons.org/licenses/by/2.5/ar http://hdl.handle.net/20.500.12110/paper_14795876_v5_n_p_vonEuw