PMID- 37107298 OWN - NLM STAT- PubMed-not-MEDLINE LR - 20230430 IS - 2076-3921 (Print) IS - 2076-3921 (Electronic) IS - 2076-3921 (Linking) VI - 12 IP - 4 DP - 2023 Apr 13 TI - KDELC2 Upregulates Glioblastoma Angiogenesis via Reactive Oxygen Species Activation and Tumor-Associated Macrophage Proliferation. LID - 10.3390/antiox12040923 [doi] LID - 923 AB - Glioblastoma is notorious for its rapid progression and neovascularization. In this study, it was found that KDEL (Lys-Asp-Glu-Leu) containing 2 (KDELC2) stimulated vasculogenic factor expression and induced human umbilical vein endothelial cell (HUVEC) proliferation. The NLRP3 inflammasome and autophagy activation via hypoxic inducible factor 1 alpha (HIF-1alpha) and mitochondrial reactive oxygen species (ROS) production was also confirmed. The application of the NLRP3 inflammasome inhibitor MCC950 and autophagy inhibitor 3-methyladenine (3-MA) indicated that the above phenomenon activation correlated with an endothelial overgrowth. Furthermore, KDELC2 suppression decreased the endoplasmic reticulum (ER) stress factors' expression. The ER stress inhibitors, such as salubrinal and GSK2606414, significantly suppressed HUVEC proliferation, indicating that ER stress promotes glioblastoma vascularization. Finally, shKDELC2 glioblastoma-conditioned medium (CM) stimulated TAM polarization and induced THP-1 cells to transform into M1 macrophages. In contrast, THP-1 cells co-cultured with compensatory overexpressed (OE)-KDELC2 glioblastoma cells increased IL-10 secretion, a biomarker of M2 macrophages. HUVECs co-cultured with shKDELC2 glioblastoma-polarized THP-1 cells were less proliferative, demonstrating that KDELC2 promotes angiogenesis. Mito-TEMPO and MCC950 increased caspase-1p20 and IL-1beta expression in THP-1 macrophages, indicating that mitochondrial ROS and autophagy could also interrupt THP-1-M1 macrophage polarization. In conclusion, mitochondrial ROS, ER stress, and the TAMs resulting from OE-KDELC2 glioblastoma cells play important roles in upregulating glioblastoma angiogenesis. FAU - Tsai, Yu-Ling AU - Tsai YL AD - Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan. FAU - Chen, Ying AU - Chen Y AUID- ORCID: 0000-0001-6303-5858 AD - Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan. FAU - Chen, Ying-Chuan AU - Chen YC AD - Department of Physiology and Biophysics, National Defense Medical Center, Taipei 114, Taiwan. FAU - Tsai, Wen-Chiuan AU - Tsai WC AD - Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan. LA - eng GR - MOST 109-2320-B-016-009/Ministry of Science and Technology/ GR - TSGH-E-110229, TSGH-E-112223/Tri-Service General Hospital/ GR - MND-MAB-D-111115/National Defense Medical Center/ PT - Journal Article DEP - 20230413 PL - Switzerland TA - Antioxidants (Basel) JT - Antioxidants (Basel, Switzerland) JID - 101668981 PMC - PMC10136350 OTO - NOTNLM OT - ER stress OT - HUVEC OT - KDELC2 OT - ROS OT - TAM OT - THP-1 OT - angiogenesis OT - glioblastoma OT - macrophage differentiation OT - tube formation COIS- The authors declare no conflict of interest. EDAT- 2023/04/28 06:41 MHDA- 2023/04/28 06:42 PMCR- 2023/04/13 CRDT- 2023/04/28 01:20 PHST- 2023/03/10 00:00 [received] PHST- 2023/04/07 00:00 [revised] PHST- 2023/04/11 00:00 [accepted] PHST- 2023/04/28 06:42 [medline] PHST- 2023/04/28 06:41 [pubmed] PHST- 2023/04/28 01:20 [entrez] PHST- 2023/04/13 00:00 [pmc-release] AID - antiox12040923 [pii] AID - antioxidants-12-00923 [pii] AID - 10.3390/antiox12040923 [doi] PST - epublish SO - Antioxidants (Basel). 2023 Apr 13;12(4):923. doi: 10.3390/antiox12040923.