File Download
Supplementary

postgraduate thesis: Harnessing single-cell transcriptomics analysis to determine cellular heterogeneity, progression, and early markers of conventional chondrosarcoma

TitleHarnessing single-cell transcriptomics analysis to determine cellular heterogeneity, progression, and early markers of conventional chondrosarcoma
Authors
Advisors
Issue Date2021
PublisherThe University of Hong Kong (Pokfulam, Hong Kong)
Citation
Su, Z. [粟澤卓]. (2021). Harnessing single-cell transcriptomics analysis to determine cellular heterogeneity, progression, and early markers of conventional chondrosarcoma. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.
AbstractChondrosarcoma is a cancer of cartilage producing cells and is relatively rare. Conventional chondrosarcomas including central (75%), peripheral (10%), periosteal (1%) variants are the most common form of chondrosarcoma that can arise from a benign tumour. However, there is currently no clear diagnostic marker for malignant transformation and clinical decision making. We used single-cell RNA sequencing (scRNA-seq) to characterize primary tumour tissues from five patients with central chondrosarcoma, one patient with a benign tumour, as well as a chondroblastic osteosarcoma and a fetal femur as controls. This atlas of 25,739 single cells was analysed to identify tumour heterogeneity, malignant transformation, and early markers of central chondrosarcoma. Based on inter-tumoral heterogeneity at single-cell resolution, we found that differentiated tumours were enriched for two distinct neoplastic cell clusters. One cluster resembled normal resting chondrocytes from the foetal femur while the other, which was subjected to cellular reprograming and endoplasmic reticulum stress (ER), indicated malignant transformation. Further single-cell analysis on additional central, peripheral, and dedifferentiated chondrosarcomas revalidated ER stress as a specific early marker for central chondrosarcoma. In addition, we revalidated the cancer development trajectory that the NF-B pathway regulated the survival of chondrosarcoma cells by alleviating ER stress-induced apoptosis. Importantly, coupled with a large cohort of bulk expression profiles, we proposed a prognostic model and identified DNA Damage Inducible Transcript 3 (DDIT3), commonly known as CHOP which mediates ER stress-induced apoptosis, as an early marker for chondrosarcoma. Overall, these findings help decipher tumour heterogeneity and malignant transformation and provide molecular signatures for the diagnosis and grading of chondrosarcoma.
DegreeDoctor of Philosophy
SubjectBones - Cancer - Diagnosis
Soft tissue tumors - Diagnosis
Cartilage cells
Dept/ProgramOrthopaedics and Traumatology
Persistent Identifierhttp://hdl.handle.net/10722/308600

 

DC FieldValueLanguage
dc.contributor.advisorCheung, SCK-
dc.contributor.advisorHo, JWK-
dc.contributor.advisorLu, WW-
dc.contributor.authorSu, Zezhuo-
dc.contributor.author粟澤卓-
dc.date.accessioned2021-12-06T01:03:58Z-
dc.date.available2021-12-06T01:03:58Z-
dc.date.issued2021-
dc.identifier.citationSu, Z. [粟澤卓]. (2021). Harnessing single-cell transcriptomics analysis to determine cellular heterogeneity, progression, and early markers of conventional chondrosarcoma. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.-
dc.identifier.urihttp://hdl.handle.net/10722/308600-
dc.description.abstractChondrosarcoma is a cancer of cartilage producing cells and is relatively rare. Conventional chondrosarcomas including central (75%), peripheral (10%), periosteal (1%) variants are the most common form of chondrosarcoma that can arise from a benign tumour. However, there is currently no clear diagnostic marker for malignant transformation and clinical decision making. We used single-cell RNA sequencing (scRNA-seq) to characterize primary tumour tissues from five patients with central chondrosarcoma, one patient with a benign tumour, as well as a chondroblastic osteosarcoma and a fetal femur as controls. This atlas of 25,739 single cells was analysed to identify tumour heterogeneity, malignant transformation, and early markers of central chondrosarcoma. Based on inter-tumoral heterogeneity at single-cell resolution, we found that differentiated tumours were enriched for two distinct neoplastic cell clusters. One cluster resembled normal resting chondrocytes from the foetal femur while the other, which was subjected to cellular reprograming and endoplasmic reticulum stress (ER), indicated malignant transformation. Further single-cell analysis on additional central, peripheral, and dedifferentiated chondrosarcomas revalidated ER stress as a specific early marker for central chondrosarcoma. In addition, we revalidated the cancer development trajectory that the NF-B pathway regulated the survival of chondrosarcoma cells by alleviating ER stress-induced apoptosis. Importantly, coupled with a large cohort of bulk expression profiles, we proposed a prognostic model and identified DNA Damage Inducible Transcript 3 (DDIT3), commonly known as CHOP which mediates ER stress-induced apoptosis, as an early marker for chondrosarcoma. Overall, these findings help decipher tumour heterogeneity and malignant transformation and provide molecular signatures for the diagnosis and grading of chondrosarcoma.-
dc.languageeng-
dc.publisherThe University of Hong Kong (Pokfulam, Hong Kong)-
dc.relation.ispartofHKU Theses Online (HKUTO)-
dc.rightsThe author retains all proprietary rights, (such as patent rights) and the right to use in future works.-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subject.lcshBones - Cancer - Diagnosis-
dc.subject.lcshSoft tissue tumors - Diagnosis-
dc.subject.lcshCartilage cells-
dc.titleHarnessing single-cell transcriptomics analysis to determine cellular heterogeneity, progression, and early markers of conventional chondrosarcoma-
dc.typePG_Thesis-
dc.description.thesisnameDoctor of Philosophy-
dc.description.thesislevelDoctoral-
dc.description.thesisdisciplineOrthopaedics and Traumatology-
dc.description.naturepublished_or_final_version-
dc.date.hkucongregation2021-
dc.identifier.mmsid991044448907503414-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats