Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma

The role of tumor necrosis factor-α (TNF-α) in shaping the tumor microenvironment is ambiguous. Consistent with its uncertain role in melanoma, TNF-α plays a dual role, either acting as a cytotoxic cytokine or favoring a tumorigenic inflammatory microenvironment. TNF-α signals via two cognate recept...

Full description

Bibliographic Details
Main Authors: Yamila I. Rodriguez, Ludmila E. Campos, Melina G. Castro, Nadia Bannoud, Ada G. Blidner, Verónica P. Filippa, Diego O. Croci, Gabriel A. Rabinovich, Sergio E. Alvarez
Format: Article
Language:English
Published: MDPI AG 2020-11-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/9/11/2469
_version_ 1797548023394336768
author Yamila I. Rodriguez
Ludmila E. Campos
Melina G. Castro
Nadia Bannoud
Ada G. Blidner
Verónica P. Filippa
Diego O. Croci
Gabriel A. Rabinovich
Sergio E. Alvarez
author_facet Yamila I. Rodriguez
Ludmila E. Campos
Melina G. Castro
Nadia Bannoud
Ada G. Blidner
Verónica P. Filippa
Diego O. Croci
Gabriel A. Rabinovich
Sergio E. Alvarez
author_sort Yamila I. Rodriguez
collection DOAJ
description The role of tumor necrosis factor-α (TNF-α) in shaping the tumor microenvironment is ambiguous. Consistent with its uncertain role in melanoma, TNF-α plays a dual role, either acting as a cytotoxic cytokine or favoring a tumorigenic inflammatory microenvironment. TNF-α signals via two cognate receptors, namely TNFR1 (p55) and TNFR2 (p75), which mediate divergent biological activities. Here, we analyzed the impact of TNFR1 deficiency in tumor progression in the B16.F1 melanoma model. Tumors developed in mice lacking TNFR1 (TNFR1 knock-out; KO) were smaller and displayed lower proliferation compared to their wild type (WT) counterpart. Moreover, TNFR1 KO mice showed reduced tumor angiogenesis. Although no evidence of spontaneous metastases was observed, conditioned media obtained from TNFR1 KO tumors increased tumor cell migration. Whereas the analysis of tumor-associated immune cell infiltrates showed similar frequency of total and M2-polarized tumor-associated macrophages (TAMs), the percentage of CD8<sup>+</sup> T cells was augmented in TNFR1 KO tumors. Indeed, functional ex vivo assays demonstrated that CD8<sup>+</sup> T cells obtained from TNFR1KO mice displayed an increased cytotoxic function. Thus, lack of TNFR1 attenuates melanoma growth by modulating tumor cell proliferation, migration, angiogenesis and CD8<sup>+</sup> T cell accumulation and activation, suggesting that interruption of TNF-TNFR1 signaling may contribute to control tumor burden.
first_indexed 2024-03-10T14:53:28Z
format Article
id doaj.art-0624a3162ac64691a87d4bdecedb176a
institution Directory Open Access Journal
issn 2073-4409
language English
last_indexed 2024-03-10T14:53:28Z
publishDate 2020-11-01
publisher MDPI AG
record_format Article
series Cells
spelling doaj.art-0624a3162ac64691a87d4bdecedb176a2023-11-20T20:47:32ZengMDPI AGCells2073-44092020-11-01911246910.3390/cells9112469Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in MelanomaYamila I. Rodriguez0Ludmila E. Campos1Melina G. Castro2Nadia Bannoud3Ada G. Blidner4Verónica P. Filippa5Diego O. Croci6Gabriel A. Rabinovich7Sergio E. Alvarez8Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, ArgentinaInstituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, ArgentinaInstituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, ArgentinaLaboratorio de Inmunopatología, Facultad de Ciencias Exactas y Naturales, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza C5500, ArgentinaLaboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, ArgentinaInstituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, ArgentinaLaboratorio de Inmunopatología, Facultad de Ciencias Exactas y Naturales, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza C5500, ArgentinaLaboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, ArgentinaInstituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, ArgentinaThe role of tumor necrosis factor-α (TNF-α) in shaping the tumor microenvironment is ambiguous. Consistent with its uncertain role in melanoma, TNF-α plays a dual role, either acting as a cytotoxic cytokine or favoring a tumorigenic inflammatory microenvironment. TNF-α signals via two cognate receptors, namely TNFR1 (p55) and TNFR2 (p75), which mediate divergent biological activities. Here, we analyzed the impact of TNFR1 deficiency in tumor progression in the B16.F1 melanoma model. Tumors developed in mice lacking TNFR1 (TNFR1 knock-out; KO) were smaller and displayed lower proliferation compared to their wild type (WT) counterpart. Moreover, TNFR1 KO mice showed reduced tumor angiogenesis. Although no evidence of spontaneous metastases was observed, conditioned media obtained from TNFR1 KO tumors increased tumor cell migration. Whereas the analysis of tumor-associated immune cell infiltrates showed similar frequency of total and M2-polarized tumor-associated macrophages (TAMs), the percentage of CD8<sup>+</sup> T cells was augmented in TNFR1 KO tumors. Indeed, functional ex vivo assays demonstrated that CD8<sup>+</sup> T cells obtained from TNFR1KO mice displayed an increased cytotoxic function. Thus, lack of TNFR1 attenuates melanoma growth by modulating tumor cell proliferation, migration, angiogenesis and CD8<sup>+</sup> T cell accumulation and activation, suggesting that interruption of TNF-TNFR1 signaling may contribute to control tumor burden.https://www.mdpi.com/2073-4409/9/11/2469tumor necrosis factor receptorB16.F1 melanomaangiogenesisimmune microenvironment
spellingShingle Yamila I. Rodriguez
Ludmila E. Campos
Melina G. Castro
Nadia Bannoud
Ada G. Blidner
Verónica P. Filippa
Diego O. Croci
Gabriel A. Rabinovich
Sergio E. Alvarez
Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
Cells
tumor necrosis factor receptor
B16.F1 melanoma
angiogenesis
immune microenvironment
title Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
title_full Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
title_fullStr Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
title_full_unstemmed Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
title_short Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8<sup>+</sup> T Cell Function in Melanoma
title_sort tumor necrosis factor receptor 1 p55 deficiency attenuates tumor growth and intratumoral angiogenesis and stimulates cd8 sup sup t cell function in melanoma
topic tumor necrosis factor receptor
B16.F1 melanoma
angiogenesis
immune microenvironment
url https://www.mdpi.com/2073-4409/9/11/2469
work_keys_str_mv AT yamilairodriguez tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT ludmilaecampos tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT melinagcastro tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT nadiabannoud tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT adagblidner tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT veronicapfilippa tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT diegoocroci tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT gabrielarabinovich tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma
AT sergioealvarez tumornecrosisfactorreceptor1p55deficiencyattenuatestumorgrowthandintratumoralangiogenesisandstimulatescd8supsuptcellfunctioninmelanoma