Skip to main content
Top

Open Access 23-04-2024 | Osteosarcoma | Research

Exploring the impact of PDGFD in osteosarcoma metastasis through single-cell sequencing analysis

Authors: Yujing Huang, Dongyan Cao, Manxue Zhang, Yue Yang, Gengming Niu, Lina Tang, Zan Shen, Zhichang Zhang, Yueqing Bai, Daliu Min, Aina He

Published in: Cellular Oncology

Login to get access

Abstract

Purpose

The overall survival rate for metastatic osteosarcoma hovers around 20%. Responses to second-line chemotherapy, targeted therapies, and immunotherapies have demonstrated limited efficacy in metastatic osteosarcoma. Our objective is to validate differentially expressed genes and signaling pathways between non-metastatic and metastatic osteosarcoma, employing single-cell RNA sequencing (scRNA-seq) and additional functional investigations. We aim to enhance comprehension of metastatic mechanisms and potentially unveil a therapeutic target.

Methods

scRNA-seq was performed on two primary osteosarcoma lesions (1 non-metastatic and 1 metastatic). Seurat package facilitated dimensionality reduction and cluster identification. Copy number variation (CNV) was predicted using InferCNV. CellChat characterized ligand-receptor-based intercellular communication networks. Differentially expressed genes underwent GO function enrichment analysis and GSEA. Validation was achieved through the GSE152048 dataset, which identified PDGFD-PDGFRB as a common ligand-receptor pair with significant contribution. Immunohistochemistry assessed PDGFD and PDGFRB expression, while multicolor immunofluorescence and flow cytometry provided insight into spatial relationships and the tumor immune microenvironment. Kaplan-Meier survival analysis compared metastasis-free survival and overall survival between high and low levels of PDGFD and PDGFRB. Manipulation of PDGFD expression in primary osteosarcoma cells examined invasion abilities and related markers.

Results

Ten clusters encompassing osteoblasts, osteoclasts, osteocytes, fibroblasts, pericytes, endothelial cells, myeloid cells, T cells, B cells, and proliferating cells were identified. Osteoblasts, osteoclasts, and osteocytes exhibited heightened CNV levels. Ligand-receptor-based communication networks exposed significant fibroblast crosstalk with other cell types, and the PDGF signaling pathway was activated in non-metastatic osteosarcoma primary lesion. These results were corroborated by the GSE152048 dataset, confirming the prominence of PDGFD-PDGFRB as a common ligand-receptor pair. Immunohistochemistry demonstrated considerably greater PDGFD expression in non-metastatic osteosarcoma tissues and organoids, correlating with extended metastasis-free and overall survival. PDGFRB expression showed no significant variation between non-metastatic and metastatic osteosarcoma, nor strong correlations with survival times. Multicolor immunofluorescence suggested co-localization of PDGFD with PDGFRB. Flow cytometry unveiled a highly immunosuppressive microenvironment in metastatic osteosarcoma. Manipulating PDGFD expression demonstrated altered invasive abilities and marker expressions in primary osteosarcoma cells from both non-metastatic and metastatic lesions.

Conclusions

scRNA-seq illuminated the activation of the PDGF signaling pathway in primary lesion of non-metastatic osteosarcoma. PDGFD displayed an inhibitory effect on osteosarcoma metastasis, likely through the suppression of the EMT signaling pathway.
Appendix
Available only for authorised users
Literature
1.
go back to reference L. Mirabello, R.J. Troisi, S.A. Savage, Osteosarcoma incidence and survival rates from 1973 to 2004: data from the Surveillance, Epidemiology, and end results program. Cancer. 115, 1531–1543 (2009)PubMedCrossRef L. Mirabello, R.J. Troisi, S.A. Savage, Osteosarcoma incidence and survival rates from 1973 to 2004: data from the Surveillance, Epidemiology, and end results program. Cancer. 115, 1531–1543 (2009)PubMedCrossRef
2.
go back to reference H. Koksal, E. Muller, E.M. Inderberg et al., Treating osteosarcoma with CAR T cells. Scand. J. Immunol. 89, e12741 (2019)PubMedCrossRef H. Koksal, E. Muller, E.M. Inderberg et al., Treating osteosarcoma with CAR T cells. Scand. J. Immunol. 89, e12741 (2019)PubMedCrossRef
3.
go back to reference M.A. Duggan, W.F. Anderson, S. Altekruse et al., The Surveillance, Epidemiology, and end results (SEER) Program and Pathology: toward strengthening the critical relationship. Am. J. Surg. Pathol. 40, e94–e102 (2016)PubMedPubMedCentralCrossRef M.A. Duggan, W.F. Anderson, S. Altekruse et al., The Surveillance, Epidemiology, and end results (SEER) Program and Pathology: toward strengthening the critical relationship. Am. J. Surg. Pathol. 40, e94–e102 (2016)PubMedPubMedCentralCrossRef
4.
go back to reference G. Bacci, A. Briccoli, M. Rocca et al., Neoadjuvant chemotherapy for osteosarcoma of the extremities with metastases at presentation: recent experience at the Rizzoli Institute in 57 patients treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide. Ann. Oncol. 14, 1126–1134 (2003)PubMedCrossRef G. Bacci, A. Briccoli, M. Rocca et al., Neoadjuvant chemotherapy for osteosarcoma of the extremities with metastases at presentation: recent experience at the Rizzoli Institute in 57 patients treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide. Ann. Oncol. 14, 1126–1134 (2003)PubMedCrossRef
5.
go back to reference A.M. Goorin, M.B. Harris, M. Bernstein et al., Phase II/III trial of etoposide and high-dose ifosfamide in newly diagnosed metastatic osteosarcoma: a pediatric oncology group trial. J. Clin. Oncol. 20, 426–433 (2002)PubMedCrossRef A.M. Goorin, M.B. Harris, M. Bernstein et al., Phase II/III trial of etoposide and high-dose ifosfamide in newly diagnosed metastatic osteosarcoma: a pediatric oncology group trial. J. Clin. Oncol. 20, 426–433 (2002)PubMedCrossRef
6.
go back to reference W.G. Ward, K. Mikaelian, F. Dorey et al., Pulmonary metastases of stage IIB extremity osteosarcoma and subsequent pulmonary metastases. J. Clin. Oncol. 12, 1849–1858 (1994)PubMedCrossRef W.G. Ward, K. Mikaelian, F. Dorey et al., Pulmonary metastases of stage IIB extremity osteosarcoma and subsequent pulmonary metastases. J. Clin. Oncol. 12, 1849–1858 (1994)PubMedCrossRef
7.
go back to reference J.C. Gentet, M. Brunat-Mentigny, M.C. Demaille et al., Ifosfamide and etoposide in childhood osteosarcoma. A phase II study of the French Society of Paediatric Oncology. Eur. J. Cancer. 33, 232–237 (1997)PubMedCrossRef J.C. Gentet, M. Brunat-Mentigny, M.C. Demaille et al., Ifosfamide and etoposide in childhood osteosarcoma. A phase II study of the French Society of Paediatric Oncology. Eur. J. Cancer. 33, 232–237 (1997)PubMedCrossRef
8.
go back to reference F. Navid, J.R. Willert, M.B. McCarville et al., Combination of gemcitabine and docetaxel in the treatment of children and young adults with refractory bone sarcoma. Cancer. 113, 419–425 (2008)PubMedCrossRef F. Navid, J.R. Willert, M.B. McCarville et al., Combination of gemcitabine and docetaxel in the treatment of children and young adults with refractory bone sarcoma. Cancer. 113, 419–425 (2008)PubMedCrossRef
9.
go back to reference E. Palmerini, R.L. Jones, E. Marchesi et al., Gemcitabine and docetaxel in relapsed and unresectable high-grade osteosarcoma and spindle cell sarcoma of bone. BMC Cancer. 16, 280 (2016)PubMedPubMedCentralCrossRef E. Palmerini, R.L. Jones, E. Marchesi et al., Gemcitabine and docetaxel in relapsed and unresectable high-grade osteosarcoma and spindle cell sarcoma of bone. BMC Cancer. 16, 280 (2016)PubMedPubMedCentralCrossRef
10.
go back to reference R.L. 3rd Saylors, K.C. Stine, J. Sullivan et al., Cyclophosphamide plus Topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J. Clin. Oncol. 19, 3463–3469 (2001)PubMedCrossRef R.L. 3rd Saylors, K.C. Stine, J. Sullivan et al., Cyclophosphamide plus Topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J. Clin. Oncol. 19, 3463–3469 (2001)PubMedCrossRef
11.
go back to reference P. Van Winkle, A. Angiolillo, M. Krailo et al., Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the children’s Cancer Group (CCG) experience. Pediatr. Blood Cancer. 44, 338–347 (2005)PubMedCrossRef P. Van Winkle, A. Angiolillo, M. Krailo et al., Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the children’s Cancer Group (CCG) experience. Pediatr. Blood Cancer. 44, 338–347 (2005)PubMedCrossRef
12.
go back to reference G. Grignani, E. Palmerini, P. Dileo et al., A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann. Oncol. 23, 508–516 (2012)PubMedCrossRef G. Grignani, E. Palmerini, P. Dileo et al., A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann. Oncol. 23, 508–516 (2012)PubMedCrossRef
13.
go back to reference G. Grignani, E. Palmerini, V. Ferraresi et al., Sorafenib and everolimus for patients with unresectable high-grade osteosarcoma progressing after standard treatment: a non-randomised phase 2 clinical trial. Lancet Oncol. 16, 98–107 (2015)PubMedCrossRef G. Grignani, E. Palmerini, V. Ferraresi et al., Sorafenib and everolimus for patients with unresectable high-grade osteosarcoma progressing after standard treatment: a non-randomised phase 2 clinical trial. Lancet Oncol. 16, 98–107 (2015)PubMedCrossRef
14.
go back to reference B. Zhu, J. Li, Q. Xie et al., Efficacy and safety of apatinib monotherapy in advanced bone and soft tissue sarcoma: an observational study. Cancer Biol. Ther. 19, 198–204 (2018)PubMedPubMedCentralCrossRef B. Zhu, J. Li, Q. Xie et al., Efficacy and safety of apatinib monotherapy in advanced bone and soft tissue sarcoma: an observational study. Cancer Biol. Ther. 19, 198–204 (2018)PubMedPubMedCentralCrossRef
15.
go back to reference H.A. Tawbi, M. Burgess, V. Bolejack et al., Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 18, 1493–1501 (2017)PubMedPubMedCentralCrossRef H.A. Tawbi, M. Burgess, V. Bolejack et al., Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 18, 1493–1501 (2017)PubMedPubMedCentralCrossRef
16.
go back to reference A. Le Cesne, P. Marec-Berard, J.Y. Blay et al., Programmed cell death 1 (PD-1) targeting in patients with advanced osteosarcomas: results from the PEMBROSARC study. Eur. J. Cancer. 119, 151–157 (2019)PubMedCrossRef A. Le Cesne, P. Marec-Berard, J.Y. Blay et al., Programmed cell death 1 (PD-1) targeting in patients with advanced osteosarcomas: results from the PEMBROSARC study. Eur. J. Cancer. 119, 151–157 (2019)PubMedCrossRef
17.
go back to reference L. Xie, J. Xu, X. Sun et al., Apatinib plus camrelizumab (anti-PD1 therapy, SHR-1210) for advanced osteosarcoma (APFAO) progressing after chemotherapy: a single-arm, open-label, phase 2 trial. J. Immunother Cancer, 8 (2020) L. Xie, J. Xu, X. Sun et al., Apatinib plus camrelizumab (anti-PD1 therapy, SHR-1210) for advanced osteosarcoma (APFAO) progressing after chemotherapy: a single-arm, open-label, phase 2 trial. J. Immunother Cancer, 8 (2020)
19.
go back to reference R. Gorlick, P. Anderson, I. Andrulis et al., Biology of childhood osteogenic sarcoma and potential targets for therapeutic development: meeting summary. Clin. Cancer Res. 9, 5442–5453 (2003)PubMed R. Gorlick, P. Anderson, I. Andrulis et al., Biology of childhood osteogenic sarcoma and potential targets for therapeutic development: meeting summary. Clin. Cancer Res. 9, 5442–5453 (2003)PubMed
20.
go back to reference J. PosthumaDeBoer, M.A. Witlox, G.J. Kaspers et al., Molecular alterations as target for therapy in metastatic osteosarcoma: a review of literature. Clin. Exp. Metastasis. 28, 493–503 (2011)PubMedPubMedCentralCrossRef J. PosthumaDeBoer, M.A. Witlox, G.J. Kaspers et al., Molecular alterations as target for therapy in metastatic osteosarcoma: a review of literature. Clin. Exp. Metastasis. 28, 493–503 (2011)PubMedPubMedCentralCrossRef
21.
go back to reference Y. Suehara, D. Alex, A. Bowman et al., Clinical genomic sequencing of Pediatric and Adult Osteosarcoma reveals distinct molecular subsets with potentially targetable alterations. Clin. Cancer Res. 25, 6346–6356 (2019)PubMedPubMedCentralCrossRef Y. Suehara, D. Alex, A. Bowman et al., Clinical genomic sequencing of Pediatric and Adult Osteosarcoma reveals distinct molecular subsets with potentially targetable alterations. Clin. Cancer Res. 25, 6346–6356 (2019)PubMedPubMedCentralCrossRef
22.
go back to reference D. Wang, X. Niu, Z. Wang et al., Multiregion sequencing reveals the genetic heterogeneity and evolutionary history of Osteosarcoma and Matched Pulmonary metastases. Cancer Res. 79, 7–20 (2019)PubMedCrossRef D. Wang, X. Niu, Z. Wang et al., Multiregion sequencing reveals the genetic heterogeneity and evolutionary history of Osteosarcoma and Matched Pulmonary metastases. Cancer Res. 79, 7–20 (2019)PubMedCrossRef
23.
go back to reference K. Mulder, A.A. Patel, W.T. Kong et al., Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity. 54, 1883–1900 (2021). e1885PubMedCrossRef K. Mulder, A.A. Patel, W.T. Kong et al., Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity. 54, 1883–1900 (2021). e1885PubMedCrossRef
24.
go back to reference I. Tirosh, B. Izar, S.M. Prakadan et al., Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 352, 189–196 (2016)PubMedPubMedCentralCrossRef I. Tirosh, B. Izar, S.M. Prakadan et al., Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 352, 189–196 (2016)PubMedPubMedCentralCrossRef
25.
go back to reference H.W. Lee, W. Chung, H.O. Lee et al., Single-cell RNA sequencing reveals the tumor microenvironment and facilitates strategic choices to circumvent treatment failure in a chemorefractory bladder cancer patient. Genome Med. 12, 47 (2020)PubMedPubMedCentralCrossRef H.W. Lee, W. Chung, H.O. Lee et al., Single-cell RNA sequencing reveals the tumor microenvironment and facilitates strategic choices to circumvent treatment failure in a chemorefractory bladder cancer patient. Genome Med. 12, 47 (2020)PubMedPubMedCentralCrossRef
26.
go back to reference M.Z. Noman, M. Hasmim, A. Lequeux et al., Improving Cancer Immunotherapy by targeting the hypoxic Tumor Microenvironment: New opportunities and challenges. Cells, 8 (2019) M.Z. Noman, M. Hasmim, A. Lequeux et al., Improving Cancer Immunotherapy by targeting the hypoxic Tumor Microenvironment: New opportunities and challenges. Cells, 8 (2019)
27.
go back to reference I. Corre, F. Verrecchia, V. Crenn et al., The Osteosarcoma Microenvironment: A Complex But Targetable Ecosystem, Cells, 9 (2020) I. Corre, F. Verrecchia, V. Crenn et al., The Osteosarcoma Microenvironment: A Complex But Targetable Ecosystem, Cells, 9 (2020)
28.
go back to reference C. Hu, C. Liu, S. Tian et al., Comprehensive analysis of prognostic tumor microenvironment-related genes in osteosarcoma patients. BMC cancer. 20, 814 (2020)PubMedPubMedCentralCrossRef C. Hu, C. Liu, S. Tian et al., Comprehensive analysis of prognostic tumor microenvironment-related genes in osteosarcoma patients. BMC cancer. 20, 814 (2020)PubMedPubMedCentralCrossRef
29.
go back to reference C. Zhang, J.H. Zheng, Z.H. Lin et al., Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of osteosarcoma. Aging. 12, 3486–3501 (2020)PubMedPubMedCentralCrossRef C. Zhang, J.H. Zheng, Z.H. Lin et al., Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of osteosarcoma. Aging. 12, 3486–3501 (2020)PubMedPubMedCentralCrossRef
30.
go back to reference Y. Han, W. Guo, T. Ren et al., Tumor-associated macrophages promote lung metastasis and induce epithelial-mesenchymal transition in osteosarcoma by activating the COX-2/STAT3 axis. Cancer Lett., 440–441 116–125 (2019) Y. Han, W. Guo, T. Ren et al., Tumor-associated macrophages promote lung metastasis and induce epithelial-mesenchymal transition in osteosarcoma by activating the COX-2/STAT3 axis. Cancer Lett., 440–441 116–125 (2019)
31.
go back to reference K. Wolf-Dennen, N. Gordon, E.S. Kleinerman, Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology. 9, 1747677 (2020)PubMedPubMedCentralCrossRef K. Wolf-Dennen, N. Gordon, E.S. Kleinerman, Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology. 9, 1747677 (2020)PubMedPubMedCentralCrossRef
32.
go back to reference P. Dhupkar, N. Gordon, J. Stewart et al., Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases. Cancer Med. 7, 2654–2664 (2018)PubMedPubMedCentralCrossRef P. Dhupkar, N. Gordon, J. Stewart et al., Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases. Cancer Med. 7, 2654–2664 (2018)PubMedPubMedCentralCrossRef
33.
go back to reference Q.K. Yang, Y.N. Su, W. Wang et al., CONUT score or/and peripheral blood CD4+/CD8 + ratio-based web dynamic nomograms to predict the individualized survival of patients with Advanced Osteosarcoma. Cancer Manag Res. 12, 4193–4208 (2020)PubMedPubMedCentralCrossRef Q.K. Yang, Y.N. Su, W. Wang et al., CONUT score or/and peripheral blood CD4+/CD8 + ratio-based web dynamic nomograms to predict the individualized survival of patients with Advanced Osteosarcoma. Cancer Manag Res. 12, 4193–4208 (2020)PubMedPubMedCentralCrossRef
34.
go back to reference Y. Yang, W. Kang, Y. Yuan et al., circ-0007707/miR-429/PDGFD Pathway Regulates the Progression of Gastric Cancer by Modulating the Immune-Gene Signature, J. Oncol., 2022 2214686 (2022) Y. Yang, W. Kang, Y. Yuan et al., circ-0007707/miR-429/PDGFD Pathway Regulates the Progression of Gastric Cancer by Modulating the Immune-Gene Signature, J. Oncol., 2022 2214686 (2022)
35.
go back to reference K. Mu, J. Fu, J. Gai et al., Genetic alterations in the neuronal development genes are associated with changes of the tumor immune microenvironment in pancreatic cancer. Ann. Pancreat. Cancer, 6 (2023) K. Mu, J. Fu, J. Gai et al., Genetic alterations in the neuronal development genes are associated with changes of the tumor immune microenvironment in pancreatic cancer. Ann. Pancreat. Cancer, 6 (2023)
36.
go back to reference K. Jin, S. Qiu, D. Jin et al., Development of prognostic signature based on immune-related genes in muscle-invasive bladder cancer: bioinformatics analysis of TCGA database. Aging (Albany NY). 13, 1859–1871 (2021)PubMedCrossRef K. Jin, S. Qiu, D. Jin et al., Development of prognostic signature based on immune-related genes in muscle-invasive bladder cancer: bioinformatics analysis of TCGA database. Aging (Albany NY). 13, 1859–1871 (2021)PubMedCrossRef
37.
go back to reference J. Hu, L. Wang, L. Li et al., A novel focal adhesion-related risk model predicts prognosis of bladder cancer -- a bioinformatic study based on TCGA and GEO database. BMC Cancer. 22, 1158 (2022)PubMedPubMedCentralCrossRef J. Hu, L. Wang, L. Li et al., A novel focal adhesion-related risk model predicts prognosis of bladder cancer -- a bioinformatic study based on TCGA and GEO database. BMC Cancer. 22, 1158 (2022)PubMedPubMedCentralCrossRef
38.
go back to reference X. Gao, J. Yang, Y. Chen, Identification of a four immune-related genes signature based on an immunogenomic landscape analysis of clear cell renal cell carcinoma. J. Cell. Physiol. 235, 9834–9850 (2020)PubMedCrossRef X. Gao, J. Yang, Y. Chen, Identification of a four immune-related genes signature based on an immunogenomic landscape analysis of clear cell renal cell carcinoma. J. Cell. Physiol. 235, 9834–9850 (2020)PubMedCrossRef
39.
go back to reference L. He, H. Yang, J. Huang, The tumor immune microenvironment and immune-related signature predict the chemotherapy response in patients with osteosarcoma. BMC Cancer. 21, 581 (2021)PubMedPubMedCentralCrossRef L. He, H. Yang, J. Huang, The tumor immune microenvironment and immune-related signature predict the chemotherapy response in patients with osteosarcoma. BMC Cancer. 21, 581 (2021)PubMedPubMedCentralCrossRef
40.
go back to reference W.J. LaRochelle, M. Jeffers, W.F. McDonald et al., PDGF-D, a new protease-activated growth factor. Nat. Cell. Biol. 3, 517–521 (2001)PubMedCrossRef W.J. LaRochelle, M. Jeffers, W.F. McDonald et al., PDGF-D, a new protease-activated growth factor. Nat. Cell. Biol. 3, 517–521 (2001)PubMedCrossRef
41.
go back to reference X. Li, U. Eriksson, Novel PDGF family members: PDGF-C and PDGF-D. Cytokine Growth Factor. Rev. 14, 91–98 (2003)PubMedCrossRef X. Li, U. Eriksson, Novel PDGF family members: PDGF-C and PDGF-D. Cytokine Growth Factor. Rev. 14, 91–98 (2003)PubMedCrossRef
42.
go back to reference E. Bergsten, M. Uutela, X. Li et al., PDGF-D is a specific, protease-activated ligand for the PDGF beta-receptor. Nat. Cell. Biol. 3, 512–516 (2001)PubMedCrossRef E. Bergsten, M. Uutela, X. Li et al., PDGF-D is a specific, protease-activated ligand for the PDGF beta-receptor. Nat. Cell. Biol. 3, 512–516 (2001)PubMedCrossRef
43.
go back to reference M. Cadamuro, S. Brivio, J. Mertens et al., Platelet-derived growth factor-D enables liver myofibroblasts to promote tumor lymphangiogenesis in cholangiocarcinoma. J. Hepatol. 70, 700–709 (2019)PubMedCrossRef M. Cadamuro, S. Brivio, J. Mertens et al., Platelet-derived growth factor-D enables liver myofibroblasts to promote tumor lymphangiogenesis in cholangiocarcinoma. J. Hepatol. 70, 700–709 (2019)PubMedCrossRef
44.
go back to reference D. Kong, S. Banerjee, W. Huang et al., Mammalian target of rapamycin repression by 3,3’-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-D-overexpressing PC3 cells. Cancer Res. 68, 1927–1934 (2008)PubMedPubMedCentralCrossRef D. Kong, S. Banerjee, W. Huang et al., Mammalian target of rapamycin repression by 3,3’-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-D-overexpressing PC3 cells. Cancer Res. 68, 1927–1934 (2008)PubMedPubMedCentralCrossRef
45.
go back to reference C.V. Ustach, M.E. Taube, N.J. Jr. Hurst et al., A potential oncogenic activity of platelet-derived growth factor d in prostate cancer progression. Cancer Res. 64, 1722–1729 (2004)PubMedPubMedCentralCrossRef C.V. Ustach, M.E. Taube, N.J. Jr. Hurst et al., A potential oncogenic activity of platelet-derived growth factor d in prostate cancer progression. Cancer Res. 64, 1722–1729 (2004)PubMedPubMedCentralCrossRef
46.
go back to reference S. Sethi, F.H. Sarkar, Q. Ahmed et al., Molecular markers of epithelial-to-mesenchymal transition are associated with tumor aggressiveness in breast carcinoma. Transl Oncol. 4, 222–226 (2011)PubMedPubMedCentralCrossRef S. Sethi, F.H. Sarkar, Q. Ahmed et al., Molecular markers of epithelial-to-mesenchymal transition are associated with tumor aggressiveness in breast carcinoma. Transl Oncol. 4, 222–226 (2011)PubMedPubMedCentralCrossRef
47.
go back to reference J. Liu, S. Liao, Y. Huang et al., PDGF-D improves drug delivery and efficacy via vascular normalization, but promotes lymphatic metastasis by activating CXCR4 in breast cancer. Clin. Cancer Res. 17, 3638–3648 (2011)PubMedPubMedCentralCrossRef J. Liu, S. Liao, Y. Huang et al., PDGF-D improves drug delivery and efficacy via vascular normalization, but promotes lymphatic metastasis by activating CXCR4 in breast cancer. Clin. Cancer Res. 17, 3638–3648 (2011)PubMedPubMedCentralCrossRef
48.
go back to reference J. Li, X. Zhi, Y. Sun et al., The PDGF Family Is Associated with Activated Tumor Stroma and Poor Prognosis in Ovarian Cancer, Dis. Markers, 2022 5940049 (2022) J. Li, X. Zhi, Y. Sun et al., The PDGF Family Is Associated with Activated Tumor Stroma and Poor Prognosis in Ovarian Cancer, Dis. Markers, 2022 5940049 (2022)
49.
go back to reference R.S. Olsen, J. Dimberg, R. Geffers et al., Possible role and therapeutic target of PDGF-D signalling in Colorectal Cancer. Cancer Invest. 37, 99–112 (2019)PubMedCrossRef R.S. Olsen, J. Dimberg, R. Geffers et al., Possible role and therapeutic target of PDGF-D signalling in Colorectal Cancer. Cancer Invest. 37, 99–112 (2019)PubMedCrossRef
50.
go back to reference H. Zhang, J.D. Sun, L.J. Yan et al., PDGF-D/PDGFRbeta promotes tongue squamous carcinoma cell (TSCC) progression via activating p38/AKT/ERK/EMT signal pathway. Biochem. Biophys. Res. Commun. 478, 845–851 (2016)PubMedCrossRef H. Zhang, J.D. Sun, L.J. Yan et al., PDGF-D/PDGFRbeta promotes tongue squamous carcinoma cell (TSCC) progression via activating p38/AKT/ERK/EMT signal pathway. Biochem. Biophys. Res. Commun. 478, 845–851 (2016)PubMedCrossRef
51.
go back to reference L. Xu, R. Tong, D.M. Cochran et al., Blocking platelet-derived growth factor-D/platelet-derived growth factor receptor beta signaling inhibits human renal cell carcinoma progression in an orthotopic mouse model. Cancer Res. 65, 5711–5719 (2005)PubMedCrossRef L. Xu, R. Tong, D.M. Cochran et al., Blocking platelet-derived growth factor-D/platelet-derived growth factor receptor beta signaling inhibits human renal cell carcinoma progression in an orthotopic mouse model. Cancer Res. 65, 5711–5719 (2005)PubMedCrossRef
53.
go back to reference E. Borkham-Kamphorst, P. Alexi, L. Tihaa et al., Platelet-derived growth factor-D modulates extracellular matrix homeostasis and remodeling through TIMP-1 induction and attenuation of MMP-2 and MMP-9 gelatinase activities. Biochem. Biophys. Res. Commun. 457, 307–313 (2015)PubMedCrossRef E. Borkham-Kamphorst, P. Alexi, L. Tihaa et al., Platelet-derived growth factor-D modulates extracellular matrix homeostasis and remodeling through TIMP-1 induction and attenuation of MMP-2 and MMP-9 gelatinase activities. Biochem. Biophys. Res. Commun. 457, 307–313 (2015)PubMedCrossRef
54.
go back to reference M. Uutela, M. Wirzenius, K. Paavonen et al., PDGF-D induces macrophage recruitment, increased interstitial pressure, and blood vessel maturation during angiogenesis. Blood. 104, 3198–3204 (2004)PubMedCrossRef M. Uutela, M. Wirzenius, K. Paavonen et al., PDGF-D induces macrophage recruitment, increased interstitial pressure, and blood vessel maturation during angiogenesis. Blood. 104, 3198–3204 (2004)PubMedCrossRef
55.
go back to reference S. Ma, T. Tang, X. Wu et al., PDGF-D-PDGFRbeta signaling enhances IL-15-mediated human natural killer cell survival. Proc. Natl. Acad. Sci. U S A, 119 (2022) S. Ma, T. Tang, X. Wu et al., PDGF-D-PDGFRbeta signaling enhances IL-15-mediated human natural killer cell survival. Proc. Natl. Acad. Sci. U S A, 119 (2022)
56.
go back to reference M. Brahmi, T. Lesluyes, A. Dufresne et al., Expression and prognostic significance of PDGF ligands and receptors across soft tissue sarcomas. ESMO Open. 6, 100037 (2021)PubMedPubMedCentralCrossRef M. Brahmi, T. Lesluyes, A. Dufresne et al., Expression and prognostic significance of PDGF ligands and receptors across soft tissue sarcomas. ESMO Open. 6, 100037 (2021)PubMedPubMedCentralCrossRef
57.
go back to reference M. Ehnman, E. Missiaglia, E. Folestad et al., Distinct effects of ligand-induced PDGFRalpha and PDGFRbeta signaling in the human rhabdomyosarcoma tumor cell and stroma cell compartments. Cancer Res. 73, 2139–2149 (2013)PubMedPubMedCentralCrossRef M. Ehnman, E. Missiaglia, E. Folestad et al., Distinct effects of ligand-induced PDGFRalpha and PDGFRbeta signaling in the human rhabdomyosarcoma tumor cell and stroma cell compartments. Cancer Res. 73, 2139–2149 (2013)PubMedPubMedCentralCrossRef
58.
go back to reference J.H. McCarty, MMP9 clears the way for metastatic cell penetration across the blood-brain barrier. Cancer Res. 83, 1167–1169 (2023)PubMedCrossRef J.H. McCarty, MMP9 clears the way for metastatic cell penetration across the blood-brain barrier. Cancer Res. 83, 1167–1169 (2023)PubMedCrossRef
59.
go back to reference J.P. Thiery, Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer. 2, 442–454 (2002)PubMedCrossRef J.P. Thiery, Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer. 2, 442–454 (2002)PubMedCrossRef
60.
go back to reference R. Roy, J. Yang, M.A. Moses, Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J. Clin. Oncol. 27, 5287–5297 (2009)PubMedPubMedCentralCrossRef R. Roy, J. Yang, M.A. Moses, Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J. Clin. Oncol. 27, 5287–5297 (2009)PubMedPubMedCentralCrossRef
61.
go back to reference M.H. Lu, M.F. Fan, X.D. Yu, NSD2 promotes osteosarcoma cell proliferation and metastasis by inhibiting E-cadherin expression. Eur. Rev. Med. Pharmacol. Sci. 21, 928–936 (2017)PubMed M.H. Lu, M.F. Fan, X.D. Yu, NSD2 promotes osteosarcoma cell proliferation and metastasis by inhibiting E-cadherin expression. Eur. Rev. Med. Pharmacol. Sci. 21, 928–936 (2017)PubMed
62.
go back to reference W. Liu, R.H. Qiao, D.M. Wang et al., UHRF1 promotes human osteosarcoma cell invasion by downregulating the expression of E–cadherin in an Rb1–dependent manner. Mol. Med. Rep. 13, 315–320 (2016)PubMedCrossRef W. Liu, R.H. Qiao, D.M. Wang et al., UHRF1 promotes human osteosarcoma cell invasion by downregulating the expression of E–cadherin in an Rb1–dependent manner. Mol. Med. Rep. 13, 315–320 (2016)PubMedCrossRef
63.
go back to reference H. Yang, Y. Zhang, Z. Zhou et al., Transcription factor Snai1-1 induces osteosarcoma invasion and metastasis by inhibiting E-cadherin expression. Oncol. Lett. 8, 193–197 (2014)PubMedPubMedCentralCrossRef H. Yang, Y. Zhang, Z. Zhou et al., Transcription factor Snai1-1 induces osteosarcoma invasion and metastasis by inhibiting E-cadherin expression. Oncol. Lett. 8, 193–197 (2014)PubMedPubMedCentralCrossRef
64.
go back to reference Q. Yuan, H. Yu, J. Chen et al., ADAM10 promotes cell growth, migration, and invasion in osteosarcoma via regulating E-cadherin/beta-catenin signaling pathway and is regulated by miR-122-5p. Cancer Cell. Int. 20, 99 (2020)PubMedPubMedCentralCrossRef Q. Yuan, H. Yu, J. Chen et al., ADAM10 promotes cell growth, migration, and invasion in osteosarcoma via regulating E-cadherin/beta-catenin signaling pathway and is regulated by miR-122-5p. Cancer Cell. Int. 20, 99 (2020)PubMedPubMedCentralCrossRef
65.
go back to reference W.H. Lin, L.M. Cooper, P.Z. Anastasiadis, Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front. Cell. Dev. Biol. 11, 1137013 (2023)PubMedPubMedCentralCrossRef W.H. Lin, L.M. Cooper, P.Z. Anastasiadis, Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front. Cell. Dev. Biol. 11, 1137013 (2023)PubMedPubMedCentralCrossRef
67.
68.
go back to reference H. Wensman, H. Goransson, K.J. Leuchowius et al., Extensive expression of craniofacial related homeobox genes in canine mammary sarcomas. Breast Cancer Res. Treat. 118, 333–343 (2009)PubMedCrossRef H. Wensman, H. Goransson, K.J. Leuchowius et al., Extensive expression of craniofacial related homeobox genes in canine mammary sarcomas. Breast Cancer Res. Treat. 118, 333–343 (2009)PubMedCrossRef
69.
go back to reference A. Shen, Y. Zhang, H. Yang et al., Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma. J. Surg. Oncol. 105, 830–834 (2012)PubMedCrossRef A. Shen, Y. Zhang, H. Yang et al., Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma. J. Surg. Oncol. 105, 830–834 (2012)PubMedCrossRef
Metadata
Title
Exploring the impact of PDGFD in osteosarcoma metastasis through single-cell sequencing analysis
Authors
Yujing Huang
Dongyan Cao
Manxue Zhang
Yue Yang
Gengming Niu
Lina Tang
Zan Shen
Zhichang Zhang
Yueqing Bai
Daliu Min
Aina He
Publication date
23-04-2024
Publisher
Springer Netherlands
Published in
Cellular Oncology
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-024-00949-3
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine