Spatial transcriptomics reveals unique metabolic profile and key oncogenic regulators of cervical squamous cell carcinoma

被引:0
|
作者
Zhou, Limin [1 ]
Liu, Jiejie [2 ,3 ]
Yao, Peipei [4 ]
Liu, Xing [2 ,3 ]
Chen, Fei [4 ]
Chen, Yu [2 ,3 ]
Zhou, Li [4 ]
Shen, Chao [5 ]
Zhou, You [6 ,7 ]
Du, Xin [1 ]
Hu, Junbo [1 ]
机构
[1] Huazhong Univ Sci & Technol, Maternal & Child Hlth Hosp Hubei Prov, Tongji Med Coll, Wuhan 430070, Peoples R China
[2] Wuhan Univ, RNA Inst, Coll Life Sci, State Key Lab Virol, Wuhan 430072, Peoples R China
[3] Wuhan Univ, RNA Inst, Frontier Sci Ctr Immunol & Metab, Wuhan 430072, Peoples R China
[4] Wuhan Univ, Inst Vaccine Res, Taikang Med Sch, Sch Basic Med Sci,Anim Biosafety Level Lab 3, Wuhan 430071, Peoples R China
[5] Wuhan Univ, Coll Life Sci, Hubei Key Lab Cell Homeostasis, Wuhan 430072, Hubei, Peoples R China
[6] Cardiff Univ, Syst Immun Res Inst, Cardiff CF14 4XN, Wales
[7] Cardiff Univ, Sch Med, Div Infect & Immun, Cardiff CF14 4XN, Wales
基金
国家重点研发计划;
关键词
Cervical squamous cell carcinoma (CSCC); Spatial transcriptomics (ST); APP; TRPS1; Tumor metabolism; TUMOR MICROENVIRONMENT; CANCER; INFLAMMATION; METASTASIS; MECHANISMS; NETWORK; ZEB1;
D O I
10.1186/s12967-024-06011-y
中图分类号
R-3 [医学研究方法]; R3 [基础医学];
学科分类号
1001 ;
摘要
BackgroundAs a prevalent and deadly malignant tumor, the treatment outcomes for late-stage patients with cervical squamous cell carcinoma (CSCC) are often suboptimal. Previous studies have shown that tumor progression is closely related with tumor metabolism and microenvironment reshaping, with disruptions in energy metabolism playing a critical role in this process. To delve deeper into the understanding of CSCC development, our research focused on analyzing the tumor microenvironment and metabolic characteristics across different regions of tumor tissue.MethodsUtilizing spatial transcriptomics (ST) sequencing technology, we conducted a study on FFPE (formalin-fixed paraffin-embedded) tumor samples from CSCC patients. Coupled with single-cell RNA sequencing (scRNA-seq) data after deconvolution, we described spatial distribution maps of tumor leading edge and core regions in detail. Tumor tissues were classified into hypermetabolic and hypometabolic regions to analyze the metabolism profiles and tumor differentiation degree across different spatial areas. We also employed The Cancer Genome Atlas (TCGA) database to examine the analysis results of ST data.ResultsOur findings indicated a more complex tumor microenvironment in hypermetabolic regions. Cell-cell communication analysis showed that various cells in tumor microenvironment were influenced by the signalling molecule APP released by cancer cells and higher expression of APP was observed in hypermetabolic regions. Furthermore, our results revealed the correlation between APP and the transcription factor TRPS1. Both APP and TRPS1 demonstrated significant effects on cancer cell proliferation, migration, and invasion, potentially contributing to tumor progression.ConclusionsUtilizing ST, scRNA-seq, and TCGA database, we examined the spatial metabolic profiles of CSCC tissues, including metabolism distribution, metabolic variations, and the relationship between metabolism and tumor differentiation degree. Additionally, potential cancer-promoting factors were proposed, offering a valuable foundation for the development of more effective treatment strategies for CSCC.
引用
收藏
页数:21
相关论文
共 50 条
  • [41] Identification of key immune cell-related genes involved in tumorigenesis and prognosis of cervical squamous cell carcinoma
    Yan, Chunxiao
    Ma, Yanyan
    Li, Junyan
    Chen, Xuejun
    Ma, Jiong
    HUMAN VACCINES & IMMUNOTHERAPEUTICS, 2023, 19 (02)
  • [42] Spatial transcriptomics and single cell sequencing identifies a unique injury associated resident macrophage subpopulation and reveals differential response to injury
    Erman, Elise
    Cheung, Matthew D.
    La Fontaine, Jennifer
    Yang, Zhengqin
    Agarwal, Anupam
    George, James
    JOURNAL OF IMMUNOLOGY, 2022, 208 (01):
  • [43] Integration of single-cell and spatial transcriptomics reveals fibroblast subtypes in hepatocellular carcinoma: spatial distribution, differentiation trajectories, and therapeutic potential
    Liu, Yue
    Dong, Guoping
    Yu, Jie
    Liang, Ping
    JOURNAL OF TRANSLATIONAL MEDICINE, 2025, 23 (01)
  • [44] Molecular characterisation in tongue squamous cell carcinoma reveals key variants potentially linked to clinical outcomes
    Alsofyani, Abeer A.
    Dallol, Ashraf
    Farraj, Suha A.
    Alsiary, Rawiah A.
    Samkari, Alaa
    Alhaj-Hussain, Baraa T.
    Khan, Jalaluddin Azam
    Al-Maghrabi, Jaudah
    Al-Khayyat, Shadi S.
    Alkhatabi, Heba
    Elaimi, Aisha
    Buhmeida, Abdelbaset
    Johargy, Ayman Khalid
    Abuzenadah, Adel M.
    Azhar, Esam I.
    Al-Qahtani, Mohammed H.
    CANCER BIOMARKERS, 2020, 28 (02) : 213 - 220
  • [45] Co-expression analysis reveals key gene modules and pathways of oral squamous cell carcinoma
    Li, Xiao
    Hu, Wei-Wei
    Wang, Li
    Yang, Xiang-Hui
    CANCER BIOMARKERS, 2018, 22 (04) : 763 - 771
  • [46] miRNA expression profile of vulvar squamous cell carcinoma and identification of the oncogenic role of miR-590-5p
    Yang, Xiuhua
    Wu, Xin
    ONCOLOGY REPORTS, 2016, 35 (01) : 398 - 408
  • [47] NMR metabolomics of human lung tumours reveals distinct metabolic signatures for adenocarcinoma and squamous cell carcinoma
    Rocha, Claudia M.
    Barros, Antonio S.
    Goodfellow, Brian J.
    Carreira, Isabel M.
    Gomes, Ana
    Sousa, Vitor
    Bernardo, Joao
    Carvalho, Lina
    Gil, Ana M.
    Duarte, Iola F.
    CARCINOGENESIS, 2015, 36 (01) : 68 - 75
  • [48] The effect of extracellular vesicles derived from oral squamous cell carcinoma on the metabolic profile of oral fibroblasts
    Lipka, Aleksandra
    Soland, Tine M.
    Nieminen, Anni I.
    Sapkota, Dipak
    Haug, Trude M.
    Galtung, Hilde K.
    FRONTIERS IN MOLECULAR BIOSCIENCES, 2025, 12
  • [49] Screening for long noncoding RNAs associated with oral squamous cell carcinoma reveals the potentially oncogenic actions of DLEU1
    Koyo Nishiyama
    Reo Maruyama
    Takeshi Niinuma
    Masahiro Kai
    Hiroshi Kitajima
    Mutsumi Toyota
    Yui Hatanaka
    Tomohiro Igarashi
    Jun-ichi Kobayashi
    Kazuhiro Ogi
    Hironari Dehari
    Akihiro Miyazaki
    Akira Yorozu
    Eiichiro Yamamoto
    Masashi Idogawa
    Yasushi Sasaki
    Tamotsu Sugai
    Takashi Tokino
    Hiroyoshi Hiratsuka
    Hiromu Suzuki
    Cell Death & Disease, 9
  • [50] Screening for long noncoding RNAs associated with oral squamous cell carcinoma reveals the potentially oncogenic actions of DLEU1
    Nishiyama, Koyo
    Maruyama, Reo
    Niinuma, Takeshi
    Kai, Masahiro
    Kitajima, Hiroshi
    Toyota, Mutsumi
    Hatanaka, Yui
    Igarashi, Tomohiro
    Kobayashi, Jun-ichi
    Ogi, Kazuhiro
    Dehari, Hironari
    Miyazaki, Akihiro
    Yorozu, Akira
    Yamamoto, Eiichiro
    Idogawa, Masashi
    Sasaki, Yasushi
    Sugai, Tamotsu
    Tokino, Takashi
    Hiratsuka, Hiroyoshi
    Suzuki, Hiromu
    CELL DEATH & DISEASE, 2018, 9