Your browser doesn't support javascript.
loading
Phosphoproteomics Reveals Selective Regulation of Signaling Pathways by Lysophosphatidic Acid Species in Macrophages.
Dietze, Raimund; Szymanski, Witold; Ojasalu, Kaire; Finkernagel, Florian; Nist, Andrea; Stiewe, Thorsten; Graumann, Johannes; Müller, Rolf.
Affiliation
  • Dietze R; Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany.
  • Szymanski W; Institute of Translational Proteomics, Biochemical Pharmacological Centre, Philipps University, 35043 Marburg, Germany.
  • Ojasalu K; Core Facility Translational Proteomics, Philipps University, 35043 Marburg, Germany.
  • Finkernagel F; Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany.
  • Nist A; Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany.
  • Stiewe T; Bioinformatics Core Facility, Philipps University, 35043 Marburg, Germany.
  • Graumann J; Genomics Core Facility, Philipps University, 35043 Marburg, Germany.
  • Müller R; Genomics Core Facility, Philipps University, 35043 Marburg, Germany.
Cells ; 13(10)2024 May 09.
Article in En | MEDLINE | ID: mdl-38786034
ABSTRACT
Lysophosphatidic acid (LPA) species, prevalent in the tumor microenvironment (TME), adversely impact various cancers. In ovarian cancer, the 180 and 204 LPA species are selectively associated with shorter relapse-free survival, indicating distinct effects on cellular signaling networks. Macrophages represent a cell type of high relevance in the TME, but the impact of LPA on these cells remains obscure. Here, we uncovered distinct LPA-species-specific responses in human monocyte-derived macrophages through unbiased phosphoproteomics, with 87 and 161 phosphosites upregulated by 204 and 180 LPA, respectively, and only 24 shared sites. Specificity was even more pronounced for downregulated phosphosites (163 versus 5 sites). Considering the high levels 204 LPA in the TME and its selective association with poor survival, this finding may hold significant implications. Pathway analysis pinpointed RHO/RAC1 GTPase signaling as the predominantly impacted target, including AHRGEF and DOCK guanine exchange factors, ARHGAP GTPase activating proteins, and regulatory protein kinases. Consistent with these findings, exposure to 204 resulted in strong alterations to the actin filament network and a consequent enhancement of macrophage migration. Moreover, 204 LPA induced p38 phosphorylation, a response not mirrored by 180 LPA, whereas the pattern for AKT was reversed. Furthermore, RNA profiling identified genes involved in cholesterol/lipid metabolism as selective targets of 204 LPA. These findings imply that the two LPA species cooperatively regulate different pathways to support functions essential for pro-tumorigenic macrophages within the TME. These include cellular survival via AKT activation and migration through RHO/RAC1 and p38 signaling.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Lysophospholipids / Signal Transduction / Proteomics / Macrophages Limits: Humans Language: En Journal: Cells Year: 2024 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Lysophospholipids / Signal Transduction / Proteomics / Macrophages Limits: Humans Language: En Journal: Cells Year: 2024 Document type: Article Affiliation country: