Abstract

Abstract Background: Rothmund-Thomson syndrome (RTS) is an autosomal recessive genetic disorder characterized by a rash that progresses to poikiloderma; small stature; skeletal anomalies; sparse hair, eyelashes, and/or eyebrows, juvenile cataracts, and an increased risk to cancer. Type 2 RTS patients with biallelic RECQL4 pathogenic variants have a significantly increased incidence of osteosarcoma. However, there is currently no available RTS model that recapitulates the bone malignancy phenotype in this disease, severely limiting the ability of researchers to explore new treatment avenues. Here, we generated RTS patient-derived induced pluripotent stem cells (iPSCs) to dissect the pathological signaling leading to RTS patient-associated osteosarcoma. Methods: iPSCs were reprogrammed from fibroblasts of four individuals from two RTS families, each with one affected RTS proband with RECQL4 mutations and one unaffected parent. These patient iPSCs were differentiated to mesenchymal stem cells (MSCs) and then to osteoblasts, the cell-of-origin of osteosarcoma. All of these MSCs and osteoblasts were subjected to transcriptome analysis by RNA-seq. Based on enriched mitochondrial respiratory gene signatures, we also systematically analyzed the enzymatic activities of the major mitochondrial respiratory protein complexes including complex I/II/III and citrate synthase enzyme activity in RTS osteoblasts by Seahorse assay. Finally, we tested the potential therapeutic effect of clinical oxidative phosphorylation complex I inhibitor IACS-010759 to treat RTS cells and dissect the pharmacological mechanisms involved. Results: RTS iPSC-derived osteoblasts showed defective osteogenic differentiation and a gain of in vitro tumorigenic ability. Transcriptome analysis of RTS osteoblasts revealed decreased bone morphogenesis gene expression but aberrantly upregulated mitochondrial complex I gene expression. Metabolic assays of RTS osteoblasts demonstrated elevated mitochondrial respiratory complex I function, increased oxidative phosphorylation, but decreased ATP production. Inhibition of mitochondrial respiratory complex I activity by IACS-010759 selectively suppressed cellular respiration and cell viability of RTS osteoblasts. Furthermore, systems analysis of IACS-010759-induced changes to RTS osteoblasts suggested that inhibition of mitochondrial complex I in RTS osteoblasts leads to enhanced cell senescence, decreased MAPK signaling and cell cycle associated genes. Conclusion: In summary, we established an RTS iPSC disease platform to dissect the pathological mechanisms involved in RTS-associated osteosarcoma. Our data suggested that mitochondrial complex I is a potential therapeutic target for RTS-associated osteosarcoma and provides future insights for clinical treatment strategies. Citation Format: An Xu, Brittany E. Jewell, Dandan Zhu, Mo-Fan Huang, Yi-Hung Chen, Linchao Lu, Ruiying Zhao, Lisa L. Wang, Dung-Fang Lee. Patient-derived iPSCs reveal pharmacologic targeting mitochondrial respiratory complex I for treating Rothmund-Thomson syndrome associated osteosarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3779.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call