1
|
Zhu K, Bai Y, Lin C, Song G, Chen Y. An energy metabolism-related signature relevant to the tumor immune microenvironment in HNSCC. Discov Oncol 2025; 16:806. [PMID: 40383838 PMCID: PMC12086135 DOI: 10.1007/s12672-025-02652-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
The importance of energy metabolism in cancer was explored by accumulating studies. Energy metabolism can affect the cellular activities of tumors. However, there is few research exploring the role of energy metabolism in tumor immune microenvironment. In this context, we constructed a novel energy metabolism-related prognostic signature containing 8 genes. The risk score calculated by the signature was analyzed to be an independent value of head and neck squamous cell carcinoma (HNSCC). We further validated the effectiveness and accuracy of our signature in The Cancer Genome Atlas Program (TCGA) cohort and Gene Expression Omnibus (GEO) cohort. Moreover, we also revealed a negative correlation between the risk score and the activity of the immune processes. Finally, we validated the function of Desmoglein 2 protein (DSG2), a risk gene in the signature, in tumor progression and found that knockdown of DSG2 remarkably suppressed the proliferation and migration of HNSCC cells, which further validated our analysis. In conclusion, the energy metabolism-related gene signature we built is a prospective biomarker of HNSCC, which can offer valuable clues for the research and development of immunotherapeutic drugs in HNSCC.
Collapse
Affiliation(s)
- Kaiyu Zhu
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Yang Bai
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
- Postdoctoral Station of Basic Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Guilin Song
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China
| | - Yifei Chen
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Wang P, Wang J, Fang Z, Chen Q, Zhang Y, Qiu X, Bao Z. Novel metabolic subtypes in IDH-mutant gliomas: implications for prognosis and therapy. BMC Cancer 2025; 25:815. [PMID: 40307749 PMCID: PMC12044917 DOI: 10.1186/s12885-025-14176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Although IDH-mutant glioma generally has a better prognosis than their IDH-wildtype counterparts, considerable prognostic heterogeneity persists among patients with the same IDH mutation. Current study has primarily focused on the different IDH statuses or grades, while the metabolic heterogeneity within IDH-mutant gliomas remains insufficiently characterized. This study aims to identify transcriptomic metabolic subtypes and associated immune microenvironment differences to better understand survival variability and potential therapeutic targets in IDH-mutant glioma. METHODS Patients with IDH-mutant gliomas were included from four public datasets (TCGA, n = 373; CGGA325, n = 167; CGGA693, n = 333; GLASS, n = 100), supplemented by 22 cases from Beijing Tiantan Hospital as an independent cohort. Consensus clustering was used to define novel metabolic subtypes. Clinical features were assessed using chi-square tests and Kaplan-Meier analysis. Metabolic profiles were characterized through enrichment analysis and GSVA; immune infiltration was analyzed using CIBERSORTx and ESTIMATE. Tumor samples from the independent cohort underwent untargeted metabolomics for validation. LASSO regression was applied to select metabolic signatures, and the CGP2014 drug library was used for drug screening. RESULTS Three metabolic subtypes (C1-C3) with distinct prognoses (p < 0.05) were identified. C1 exhibited enhanced carbohydrate and nucleotide metabolism; C2 displayed upregulated amino acid and lipid metabolism; and C3 demonstrated elevated lipid, nucleotide, and vitamin metabolism. These patterns were validated in the independent cohort. Subtypes were also correlated with immune infiltration. A 13-gene metabolic signature was established to stratify prognostic risk and suggest subtype-specific drug sensitivities. CONCLUSIONS Our study provided a novel metabolic subtype for IDH-mutant glioma and highlighted these patients' metabolic heterogeneity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiayi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng Fang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qiaodong Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Xiaoguang Qiu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Wang B, Huang C, Liu X, Liu Z, Zhang Y, Zhao W, Xu Q, Ho PC, Xiao Z. iMetAct: An integrated systematic inference of metabolic activity for dissecting tumor metabolic preference and tumor-immune microenvironment. Cell Rep 2025; 44:115375. [PMID: 40053454 DOI: 10.1016/j.celrep.2025.115375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/03/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
Metabolic enzymes play a central role in cancer metabolic reprogramming, and their dysregulation creates vulnerabilities that can be exploited for therapy. However, accurately measuring metabolic enzyme activity in a high-throughput manner remains challenging due to the complex, multi-layered regulatory mechanisms involved. Here, we present iMetAct, a framework that integrates metabolic-transcription networks with an information propagation strategy to infer enzyme activity from gene expression data. iMetAct outperforms expression-based methods in predicting metabolite conversion rates by accounting for the effects of post-translational modifications. With iMetAct, we identify clinically significant subtypes of hepatocellular carcinoma with distinct metabolic preferences driven by dysregulated enzymes and metabolic regulators acting at both the transcriptional and non-transcriptional levels. Moreover, applying iMetAct to single-cell RNA sequencing data allows for the exploration of cancer cell metabolism and its interplay with immune regulation in the tumor microenvironment. An accompanying online platform further facilitates tumor metabolic analysis, patient stratification, and immune microenvironment characterization.
Collapse
Affiliation(s)
- Binxian Wang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chao Huang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xuan Liu
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhenni Liu
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yilei Zhang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Epalinges, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Zhengtao Xiao
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
4
|
Li X, Zhao Z, Cheng Y, Yan J, Ren F, Jia Y, Li J, Wang B, Liu J, Wang C, Gao M, Gu H, Fan M, Shi H, Ji M, Zhao Q. Single-cell transcriptomic analyses reveal heterogeneity and key subsets associated with survival and response to PD-1 blockade in cervical squamous cell carcinoma. Cancer Cell Int 2025; 25:90. [PMID: 40082876 PMCID: PMC11907857 DOI: 10.1186/s12935-025-03725-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Understanding the intricate tumor microenvironment (TME) is crucial for elucidating the mechanisms underlying the progression of cervical squamous cell carcinoma (CSCC) and its response to anti-PD-1 therapy. METHODS In this study, we characterized 50,649 cells obtained from the CSCC for single-cell RNA sequencing and integrated bulk sequencing data from The Cancer Genome Atlas (TCGA) and clinical samples to explore their cell composition, metabolic processes, signaling pathways, specific transcription factors, lineage tracking and response to immunotherapy. In vivo experiments were performed to validate the function of key cell subsets. RESULTS We identified ten major cell type and 35 subsets of stromal and immune cells in TME and observed distinct patterns in the metabolic processes and signaling pathways of these cells between tumor and normal tissues. Furthermore, PCNA clamp-associated factor (PCLAF)+ tumor-associated epithelial cell (TAEpis) was negatively correlated with the number of C-X-C motif chemokine ligand 13 (CXCL13)+ CD8+ T cells, overall survival, and response to anti-programmed cell death-1(PD-1) therapy in patients with CSCC. Both in vivo and in vitro experiments demonstrated that PCLAF+ TAEpis promotes the apoptosis of CD8+ T and tumor growth, while also inhibiting T cell infiltration and function. CONCLUSION Our findings illuminate the heterogeneity of the complex TME in CSCC and offer evidence supporting PCLAF+ TAEpis as a promising therapeutic target.
Collapse
Affiliation(s)
- Xia Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Zhao Zhao
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yanmei Cheng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Jiaqin Yan
- Department of Oncology, the first affiliated hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Fang Ren
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yanyan Jia
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Juanhua Li
- Department of Digestive System, Henan Electric Power Hospital, Zhengzhou, Henan Province, 450052, China
| | - Binhui Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Chenyin Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Meimei Gao
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Mingliang Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Huirong Shi
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Mei Ji
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Qitai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
5
|
Miao J, Chen B, Zhang L, Lu Z, Wang R, Wang C, Jiang X, Shen Q, Li Y, Shi D, Ouyang Y, Chen X, Deng X, Zhang S, Zou H, Chen S. Metabolic expression profiling analysis reveals pyruvate-mediated EPHB2 upregulation promotes lymphatic metastasis in head and neck squamous cell carcinomas. J Transl Med 2025; 23:316. [PMID: 40075431 PMCID: PMC11899055 DOI: 10.1186/s12967-025-06305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Lymphatic metastasis is a well-known factor for initiating distant metastasis of head and neck squamous cell carcinoma (HNSCC), which caused major death in most patients with cancer. Meanwhile, metabolic reprogramming to support metastasis is regarded as a prominent hallmark of cancers. However, how metabolic disorders drive in HNSCC remains unclear. We firstly established a new classification of HNSCC patients based on metabolism gene expression profiles from the TCGA and GEO database, and identified an enriched carbohydrate metabolism subgroup which was significantly associated with lymphatic metastasis and worse clinical outcome. Moreover, we found that highly activated pyruvate metabolism endowed tumors with EPHB2 upregulation and promoted tumor lymphangiogenesis independently of VEGF-C/VEGFR3 signaling pathway. Mechanically, high nuclear acetyl-CoA production from pyruvate metabolism promoted histone acetylation, which in turn transcriptionally upregulated EPHB2 expression and secretion in tumor cells. EPHB2 bound with EFNB1 in lymphatic endothelial cells promoted YAP/TAZ cytoplasmic retention, which alleviated YAP/TAZ-mediated prospero homeobox protein 1 (PROX1) transcriptional repression, and then triggered tumor lymphangiogenesis. Importantly, combined treatment with EFNB1-Fc and VEGFR3 inhibitor synergistic abrogated lymphangiogenesis in vitro and in vivo, suggesting that targeting EPHB2 might be a potential strategy to patients with no or slight response to VEGFR3 inhibitor. These findings uncover the mechanism by which pyruvate metabolism is linked to lymphatic metastasis of tumor and provides a promising therapeutic strategy for the prevention of HNSCC metastasis.
Collapse
Affiliation(s)
- Jingjing Miao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Boyu Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Lu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zhongming Lu
- Department of Otolaryngology Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, P. R. China
| | - Rui Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chunyang Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Xingyu Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qi Shen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Zhejiang, 311402, P. R. China
| | - Yue Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Dongni Shi
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ying Ouyang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiangfu Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaowu Deng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Siyi Zhang
- Department of Otolaryngology Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, P. R. China.
| | - Hequn Zou
- Medical School, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China.
| | - Shuwei Chen
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
6
|
Bathe OF. Tumor metabolism as a factor affecting diversity in cancer cachexia. Am J Physiol Cell Physiol 2025; 328:C908-C920. [PMID: 39870605 DOI: 10.1152/ajpcell.00677.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
Cancer cachexia is a multifaceted metabolic syndrome characterized by muscle wasting, fat redistribution, and metabolic dysregulation, commonly associated with advanced cancer but sometimes also evident in early-stage disease. More subtle body composition changes have also been reported in association with cancer, including sarcopenia, myosteatosis, and increased fat radiodensity. Emerging evidence reveals that body composition changes including sarcopenia, myosteatosis, and increased fat radiodensity, arise from distinct biological mechanisms and significantly impact survival outcomes. Importantly, these features often occur independently, with their combined presence exacerbating poor prognoses. Tumor plays a pivotal role in driving these host changes, either by acting as a metabolic parasite or by releasing mediators that disrupt normal tissue function. This review explores the diversity of tumor metabolism. It highlights the potential for tumor-specific metabolic phenotypes to influence systemic effects, including fat redistribution and sarcopenia. Addressing this tumor-host metabolic interplay requires personalized approaches that disrupt tumor metabolism while preserving host health. Promising strategies include targeted pharmacological interventions and anticachexia agents like growth differentiation factor 15 (GDF-15) inhibitors. Nutritional modifications such as ketogenic diets and omega-3 fatty acid supplementation also merit further investigation. In addition to preserving muscle, these therapies will need to be evaluated for their capability to improve survival and quality of life. This review underscores the need for further research into tumor-driven metabolic effects on the host and the development of integrative treatment strategies to address the interconnected challenges of cancer progression and cachexia.
Collapse
Affiliation(s)
- Oliver F Bathe
- Department of Surgery and Oncology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Wang Y, Qiu X, Li Q, Qin J, Ye L, Zhang X, Huang X, Wen X, Wang Z, He W, Di Y, Zhou Q. Single-cell and spatial-resolved profiling reveals cancer-associated fibroblast heterogeneity in colorectal cancer metabolic subtypes. J Transl Med 2025; 23:175. [PMID: 39934919 PMCID: PMC11817247 DOI: 10.1186/s12967-025-06103-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) presents significant treatment challenges due to its high heterogeneity and complex intercellular interactions. Further exploration of CRC subtypes and interactions among tumor-specific clusters will facilitate the development of personalized treatment strategies. METHODS Single-cell RNA sequencing and bulk RNA sequencing datasets were integrated to determine CRC metabolic subtypes by hierarchical clustering. The analysis was further extended to cellchat, pseudotime, immune infiltration, and clinicopathological relevance to explore the characteristics of secreted frizzled related protein 2 (SFRP2) + cancer-associated fibroblast (CAF) clusters, and validated by spatial transcriptomics (ST), in vivo experiments, and multiple immunohistochemistry (mIHC). RESULTS CRC samples were stably classified into three heterogeneous metabolic subtypes, each exhibiting different microenvironment and CAF heterogeneity, particularly in the distribution of SFRP2 + CAF, which was aligned with metabolic activity. SFRP2 + CAF exhibits high extracellular matrix (ECM) activity and is closely involved in cellular communication, not only promoting the malignant progression of cancer cells but also inducing the differentiation of Tregs. Compared to responders of chemotherapy, the proportion of SFRP2 + CAFs is significantly increased in non-responders. Importantly, mIHC and ST analyses confirm that cancer cells with low expression of agmatinase (AGMAT) can recruit SFRP2 + CAFs, and Treg infiltration surrounding SFRP2 + CAFs was observed. AGMAT combined with oxaliplatin showed the best efficacy in vivo, which may be associated with the inhibition of SFRP2 + CAF infiltration. CONCLUSIONS Our study identified and described the potential protumor biological properties of SFRP2 + CAFs, and AGMAT may be a valuable target for disrupting their properties.
Collapse
Affiliation(s)
- Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Xingfeng Qiu
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China
| | - Qinghai Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Lvlan Ye
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Xingxiang Huang
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China
| | - Xiangqiong Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China.
| | - Yuqin Di
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
| | - Qi Zhou
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Department of General Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou, Guangdong, 516081, China.
| |
Collapse
|
8
|
Tan W, Dai F, Ci Q, Deng Z, Liu H, Cheng Y. Characterization of tumor prognosis and sensitive chemotherapy drugs based on cuproptosis-related gene signature in ovarian cancer. BMC Womens Health 2025; 25:37. [PMID: 39849417 PMCID: PMC11761216 DOI: 10.1186/s12905-024-03519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Cuproptosis is a novel form of cell death, acting on the tricarboxylic acid cycle in mitochondrial respiration and mediated by protein lipoylation. Other cancer cell death processes, such as necroptosis, pyroptosis, and ferroptosis, have been shown to play crucial roles in the therapy and prognosis of ovarian cancer. However, the role of cuproptosis in ovarian cancer remains unclear. METHODS The expression profiles of 10 cuproptosis-related genes were extracted from GSE140082. Kaplan-Meier survival and Cox proportional hazards regression were used to identify prognostic genes for constructing risk models. Following this, Least Absolute Shrinkage and Selection Operator regression was employed to construct a risk score model. Next, a nomogram was constructed to predict overall survival in ovarian cancer. Ultimately, our analysis compared the two groups across various dimensions, including clinical characteristics, tumor progression, metabolism-related pathways, immune landscape, and drug sensitivity. RESULTS MTF1 and LIAS were identified as protective factors in ovarian cancer, with patients in the higher risk group being significantly associated with poorer survival. Furthermore, integrating the risk score with clinical characteristics in the nomogram demonstrated high specificity and sensitivity in predicting survival. A higher propotion of M2 macrophages, follicular helper T cells, and resting mast cells was observed in the high-risk group. Additionally, the IC50 values of Dasatinib, Bortezomib, Parthenolide, and Imatinib were significantly lower in the high-risk group. CONCLUSIONS The study highlights the prognostic significance of cuproptosis-related genes and provides new insights into developing pharmacological therapeutic strategies targeting cuproptosis for the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qinyu Ci
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
9
|
Li G, Cui J, Li T, Li W, Chen P. A risk signature constructed by Tregs-related genes predict the clinical outcomes and immune therapeutic response in kidney cancer. Discov Oncol 2025; 16:64. [PMID: 39833617 PMCID: PMC11747013 DOI: 10.1007/s12672-025-01787-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Regulatory T cells (Tregs) have been found to be related to immune therapeutic resistance in kidney cancer. However, the potential Tregs-related genes still need to be explored. Our study found that patients with high Tregs activity show poor prognosis. Through co-expression and differential expression analysis, we screened several Tregs-related genes (KTRGs) in kidney renal clear cell carcinoma. We further conducted the univariate Cox regression analysis and determined the prognosis-related KTRGs. Through the machine learning algorithm-Boruta, the potentially important KTRGs were screened further and submitted to construct a risk model. The risk model could predict the prognosis of RCC patients well, high risk patients show a poorer outcomes than low risk patients. Multivariate Cox regression analysis reveals that risk score is an independent prognostic factor. Then, the nomogram model based on KTRG risk score and other clinical variables was further established, which shows a high predicted accuracy and clinical benefit based on model validation methods. In addition, we found EMT, JAK/STAT3, and immune-related pathways highly enriched in high risk groups, while metabolism-related pathways show a low enrichment. Through analyzing two other external immune therapeutic datasets, we found that the risk score could predict the patient's immune therapeutic response. High-risk groups represent a worse therapeutic response than low-risk groups. In summary, we identified several Tregs-related genes and constructed a risk model to predict prognosis and immune therapeutic response. We hope these organized data can provide a theoretical basis for exploring potential Tregs' targets to synergize the immune therapy for RCC patients.
Collapse
Affiliation(s)
- Gang Li
- Department of Urology, Tangshan Gongren Hospital, 27 Wenhua Road, Tangshan, 063000, Hebei, People's Republic of China
| | - Jingmin Cui
- Department of Urology, Tangshan Gongren Hospital, 27 Wenhua Road, Tangshan, 063000, Hebei, People's Republic of China
| | - Tao Li
- Department of Urology, Tangshan Gongren Hospital, 27 Wenhua Road, Tangshan, 063000, Hebei, People's Republic of China
| | - Wenhan Li
- Department of Urology, Tangshan Gongren Hospital, 27 Wenhua Road, Tangshan, 063000, Hebei, People's Republic of China
| | - Peilin Chen
- Department of Urology, Tangshan Gongren Hospital, 27 Wenhua Road, Tangshan, 063000, Hebei, People's Republic of China.
| |
Collapse
|
10
|
Kiesworo K, Agius T, Macarthur MR, Lambelet M, Lyon A, Zhang J, Turiel G, Fan Z, d’Almeida S, Uygun K, Yeh H, Déglise S, de Bock K, Mitchell SJ, Ocampo A, Allagnat F, Longchamp A. Nicotinamide mononucleotide restores impaired metabolism, endothelial cell proliferation and angiogenesis in old sedentary male mice. iScience 2025; 28:111656. [PMID: 39868046 PMCID: PMC11763620 DOI: 10.1016/j.isci.2024.111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Aging is accompanied by a decline in neovascularization potential and increased susceptibility to ischemic injury. Here, we confirm the age-related impaired neovascularization following ischemic leg injury and impaired angiogenesis. The age-related deficits in angiogenesis arose primarily from diminished EC proliferation capacity, but not migration or VEGF sensitivity. Aged EC harvested from the mouse skeletal muscle displayed a pro-angiogenic gene expression phenotype, along with considerable changes in metabolic genes. Metabolomics analysis and 13C glucose tracing revealed impaired ATP production and blockade in glycolysis and TCA cycle in late passage HUVECs, which occurred at nicotinamide adenine dinucleotide (NAD⁺)-dependent steps, along with NAD+ depletion. Supplementation with nicotinamide mononucleotide (NMN), a precursor of NAD⁺, enhances late-passage EC proliferation and sprouting angiogenesis from aged mice aortas. Taken together, our study illustrates the importance of NAD+-dependent metabolism in the maintenance of EC proliferation capacity with age, and the therapeutic potential of NAD precursors.
Collapse
Affiliation(s)
- Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Michael R. Macarthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Martine Lambelet
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Arnaud Lyon
- Transplantation Centre and Transplantation Immunopathology Laboratory, Department of Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Guillermo Turiel
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Zheng Fan
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Korkut Uygun
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Katrien de Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sarah J. Mitchell
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, Lausanne University (UNIL), Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Indraccolo S, Signoriello S, Piga I, Esposito G, Ferrarini F, Boscolo Bragadin A, Salutari V, Pisano C, Califano D, Bignotti E, Tognon G, Simeon V, Artioli G, Ferrero A, Cinieri S, Bologna A, Chiodini P, Scognamiglio G, Bottoni C, Spina A, Russo D, Arenare L, Perrone F, Pignata S. Impact of metabolism-related markers on outcomes in ovarian cancer patients: Findings of the MITO16A/MaNGO-OV2 trial. Int J Biol Markers 2024; 39:328-337. [PMID: 39513196 DOI: 10.1177/03936155241296164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
INTRODUCTION In ovarian cancer, expression of metabolism-related markers has been investigated in several studies focusing on individual markers; however, a parallel quantitative evaluation of markers mapping to distinct metabolic processes and their prognostic value in large patient cohorts is still lacking. METHODS Here, by using immunohistochemistry followed by digital pathology, we investigated the expression of several markers related to glycolysis including monocarboxylate transporter 1 and 4 (MCT1, MCT4), glutamine metabolism (glutaminase, GLS) and hypoxia/acidosis (carbonic anhydrase 9, CA IX) in tissue microarrays of > 300 patients recruited in the MITO16A clinical trial, which involved treatment of ovarian cancer patients with carboplatin/taxol plus bevacizumab. RESULTS Regarding the prognostic impact of these markers, results indicate that GLS expression correlated with progression-free survival, but this effect disappeared when data were corrected for multiple testing. All other markers showed no correlation with clinical outcome. CONCLUSION These results indicate marked heterogeneity of expression of metabolism-associated markers in ovarian cancer; however, there was a lack of association with clinical benefit after chemotherapy/anti-vascular endothelial growth factor treatment. Notwithstanding the lack of prognostic value, knowledge of the pattern of expression of these biomarkers in tumors can be useful for patient stratification purposes when new drugs targeting these metabolic pathways will be tested.
Collapse
Affiliation(s)
- Stefano Indraccolo
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Simona Signoriello
- Medical Statistics Unit, Department of Physical and Mental Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Ilaria Piga
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Giovanni Esposito
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Federica Ferrarini
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Vanda Salutari
- Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy, Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Roma, Italy
| | - Carmela Pisano
- Uro-Gynecological Medical Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Daniela Califano
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Eliana Bignotti
- ASST Spedali Civili di Brescia, Università di Brescia, Brescia, Italy
| | - Germana Tognon
- ASST Spedali Civili di Brescia, Università di Brescia, Brescia, Italy
| | - Vittorio Simeon
- Medical Statistics Unit, Department of Physical and Mental Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Grazia Artioli
- Oncologia Medica, ULSS 2 Marca Trevigiana, Treviso, Italy
| | - Annamaria Ferrero
- Academic Division of Gynecology and Obstetrics, Mauriziano Hospital and University of Torino, Torino, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | | | - Paolo Chiodini
- Medical Statistics Unit, Department of Physical and Mental Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Giosuè Scognamiglio
- Scientific Directorate, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Carolina Bottoni
- Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy, Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Roma, Italy
| | - Anna Spina
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Daniela Russo
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Laura Arenare
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Francesco Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Sandro Pignata
- Uro-Gynecological Medical Oncology, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
12
|
Ge H, Malsiu F, Gao Y, Losmanova T, Blank F, Ott J, Medová M, Peng RW, Deng H, Dorn P, Marti TM. Inhibition of LDHB suppresses the metastatic potential of lung cancer by reducing mitochondrial GSH catabolism. Cancer Lett 2024; 611:217353. [PMID: 39615645 DOI: 10.1016/j.canlet.2024.217353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024]
Abstract
Metastasis, the leading cause of cancer death, is closely linked to lactate metabolism. Our study aimed to investigate the role of lactate dehydrogenase B (LDHB), which mainly catalyzes the conversion of lactate to pyruvate, in the metastatic potential of lung cancer. We found that LDHB silencing reduced the invasion and migration ability of lung cancer cells in vitro. On the molecular level, LDHB silencing decreased the total intracellular levels of the antioxidant glutathione (GSH). Surprisingly, LDHB silencing did not increase cellular or mitochondrial reactive oxygen species (ROS) levels. Furthermore, supplementation with GSH monoethyl ester (GSH-mee), a cell-permeable derivative of GSH, partially restored the reduced in vitro colony formation capacity, the oxygen consumption rate, and the invasion and migration capacity of lung cancer cells after LDHB silencing. Using metabolic inhibitors, we showed that the rescue of colony formation after silencing LDHB by GSH-mee was due to enhanced GSH catabolism by γ-L-Glutamyl transpeptidase (GGT), which was mainly present in the mitochondrial fraction of lung cancer cells. Furthermore, we observed that high GGT expression was a prerequisite for the rescue of migratory capacity by GSH-mee after LDHB silencing. Finally, our in vivo experiments demonstrated that targeting LDHB reduced the metastasis of human and mouse lung cancer cells in immunodeficient and immunocompetent mouse models, respectively. In conclusion, LDHB silencing decreases GSH catabolism mediated by GGT, which is primarily located in the mitochondria of cancer cells. Therefore, targeting LDHB is a promising therapeutic approach for the prevention and treatment of metastatic lung cancer.
Collapse
Affiliation(s)
- Huixiang Ge
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Fatlind Malsiu
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Tereza Losmanova
- Institute of Tissue Medicine and Pathology, ITMP, University of Bern, Bern, Switzerland
| | - Fabian Blank
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Julien Ott
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Michaela Medová
- Department for BioMedical Research, University of Bern, Bern, Switzerland; Department of Radiation Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
13
|
Xu M, Ai H, Wang D, Wang X. Gene clusters-based pathway enrichment analysis identifies four pan-cancer subtypes with distinct molecular and clinical features. Funct Integr Genomics 2024; 24:224. [PMID: 39607532 DOI: 10.1007/s10142-024-01501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Pathways-based clustering methods have been proposed to explore tumor heterogeneity. However, such methods are currently disadvantageous in that specific pathways need to be explicitly claimed. We developed the PathClustNet algorithm, a pathway-based clustering method designed to identify cancer subtypes. This method first detects gene clusters and identifies overrepresented pathways associated with them. Based on the pathway enrichment scores, it reveals cancer subtypes by clustering analysis. We applied the method to TCGA pan-cancer data and identified four pan-cancer subtypes, termed C1, C2, C3 and C4. C1 exhibited high metabolic activity, favorable survival, and the lowest TP53 mutation rate. C2 had high immune, developmental, and stromal pathway activities, the lowest tumor purity, and intratumor heterogeneity. C3, which overexpressed cell cycle and DNA repair pathways, was the most genomically unstable and had the highest TP53 mutation rate. C4 overrepresented neuronal pathways, with the lowest response rate to chemotherapy, but the highest tumor purity and genomic stability. Furthermore, age showed positive correlations with most pathways but a negative correlation with neuronal pathways. Smoking, viral infections, and alcohol use were found to affect the activities of neuron, cell cycle, immune, stromal, developmental, and metabolic pathway in varying degrees. The PathClustNet algorithm unveils a novel classification of pan-cancer based on metabolic, immune, stromal, developmental, cell cycle, and neuronal pathways. These subtypes display different molecular and clinical features to warrant the investigation of precision oncology.
Collapse
Affiliation(s)
- Mengli Xu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Hongjing Ai
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Danni Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, 211198, Nanjing, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, 211198, Nanjing, China.
- Big Data Research Institute, China Pharmaceutical University, 211198, Nanjing, China.
| |
Collapse
|
14
|
Yan H, Mao AW, Li D, Pérez-Baena MJ, Jiménez-Navas A, Wang D, Hong R, Cai W, Pérez-Losada J, Jen KY, Wang S, Peng S, Barcellos-Hoff MH, Mao JH, Fu Y, Iczkowski KA, Gulati S, Chang H. AI-Powered cellular morphometric biomarkers discovered in needle biopsy of prostatic cancer predict neoadjuvant androgen deprivation therapy response and prognosis: an international multicenter retrospective study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.17.24317411. [PMID: 39606414 PMCID: PMC11601692 DOI: 10.1101/2024.11.17.24317411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
It is imperative to identify patients with prostate cancer (PCa) who will benefit from androgen receptor signaling inhibitors that can impact quality of life upon prolonged use. Using our extensively-validated artificial-intelligence technique: cellular morphometric biomarker via machine learning (CMB-ML), we identified 13 CMBs from whole slide images of needle biopsies from the trial specimens ( NCT02430480 , n=37) that accurately predicted response to neoadjuvant androgen deprivation therapy (NADT) (AUC: 0.980). Notably, 13-CMB model stratified PCa patients into responder and non-responder groups after NADT treatment in an independent hospital cohort (n=122) that significantly associated with pathologic complete response (p=0.0005), biochemical-recurrence-free survival (p=0.024) and mTOR signaling pathway (p=0.03), suggesting potentially more clinical benefit from mTOR inhibitors in non-responder group. Additionally, genetic and genomic analysis revealed interplay between genetic variants and CMBs on NADT resistance, and provided molecular annotations for CMBs. Overall, prospective clinical implementation of 13-CMB model could assist precision care of PCa patients. Significance We describe a highly accurate CMB model to predict the therapeutic benefit in prostate cancer patients and uncover the complex interplay between genetic variants and CMBs on NADT resistance. Our model relies only on widely available needle biopsy specimens and provides a robust and cost-effective solution for clinical implementation.
Collapse
|
15
|
Weistuch C, Murgas KA, Zhu J, Norton L, Dill KA, Tannenbaum AR, Deasy JO. Normal tissue transcriptional signatures for tumor-type-agnostic phenotype prediction. Sci Rep 2024; 14:27230. [PMID: 39516498 PMCID: PMC11549333 DOI: 10.1038/s41598-024-76625-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer transcriptional patterns reflect both unique features and shared hallmarks across diverse cancer types, but whether differences in these patterns are sufficient to characterize the full breadth of tumor phenotype heterogeneity remains an open question. We hypothesized that these shared transcriptomic signatures reflect repurposed versions of functional tasks performed by normal tissues. Starting with normal tissue transcriptomic profiles, we use non-negative matrix factorization to derive six distinct transcriptomic phenotypes, called archetypes, which combine to describe both normal tissue patterns and variations across a broad spectrum of malignancies. We show that differential enrichment of these signatures correlates with key tumor characteristics, including overall patient survival and drug sensitivity, independent of clinically actionable DNA alterations. Additionally, we show that in HR+/HER2- breast cancers, metastatic tumors adopt transcriptomic signatures consistent with the invaded tissue. Broadly, our findings suggest that cancer often arrogates normal tissue transcriptomic characteristics as a component of both malignant progression and drug response. This quantitative framework provides a strategy for connecting the diversity of cancer phenotypes and could potentially help manage individual patients.
Collapse
Affiliation(s)
- Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Kevin A Murgas
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Jiening Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Ken A Dill
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, USA
| | - Allen R Tannenbaum
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, USA
- Department of Computer Science, Stony Brook University, Stony Brook, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
16
|
Zhou Z, Dong D, Yuan Y, Luo J, Liu XD, Chen LY, Wang G, Yin Y. Single cell atlas reveals multilayered metabolic heterogeneity across tumour types. EBioMedicine 2024; 109:105389. [PMID: 39393173 DOI: 10.1016/j.ebiom.2024.105389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Metabolic reprogramming plays a pivotal role in cancer progression, contributing to substantial intratumour heterogeneity and influencing tumour behaviour. However, a systematic characterization of metabolic heterogeneity across multiple cancer types at the single-cell level remains limited. METHODS We integrated 296 tumour and normal samples spanning six common cancer types to construct a single-cell compendium of metabolic gene expression profiles and identify cell type-specific metabolic properties and reprogramming patterns. A computational approach based on non-negative matrix factorization (NMF) was utilised to identify metabolic meta-programs (MMPs) showing intratumour heterogeneity. In-vitro cell experiments were conducted to confirm the associations between MMPs and chemotherapy resistance, as well as the function of key metabolic regulators. Survival analyses were performed to assess clinical relevance of cellular metabolic properties. FINDINGS Our analysis revealed shared glycolysis upregulation and divergent regulation of citric acid cycle across different cell types. In malignant cells, we identified a colorectal cancer-specific MMP associated with resistance to the cuproptosis inducer elesclomol, validated through in-vitro cell experiments. Furthermore, our findings enabled the stratification of patients into distinct prognostic subtypes based on metabolic properties of specific cell types, such as myeloid cells. INTERPRETATION This study presents a nuanced understanding of multilayered metabolic heterogeneity, offering valuable insights into potential personalized therapies targeting tumour metabolism. FUNDING National Key Research and Development Program of China (2021YFA1300601). National Natural Science Foundation of China (key grants 82030081 and 81874235). The Shenzhen High-level Hospital Construction Fund and Shenzhen Basic Research Key Project (JCYJ20220818102811024). The Lam Chung Nin Foundation for Systems Biomedicine.
Collapse
Affiliation(s)
- Zhe Zhou
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Di Dong
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Juan Luo
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiao-Ding Liu
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Long-Yun Chen
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
17
|
Deng M, Liu J, Zhang L, Lou Y, Qiu Y. Identification of molecular subtypes based on bile acid metabolism in cholangiocarcinoma. BMC Cancer 2024; 24:1313. [PMID: 39455933 PMCID: PMC11515294 DOI: 10.1186/s12885-024-13081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cholangiocarcinoma is a highly heterogeneous tumor with bile acid metabolism involving in its development. The aim of this study was to characterize bile acid metabolism and identify specific subtypes to better stratify cholangiocarcinoma patients for individualized treatment and prognostic assessment. METHODS A total of 30 bile acids were quantified using the ultra-performance liquid chromatography tandem mass spectrometry. Using Consensus clustering, the molecular subtypes related to bile acid metabolism were identified. The prognosis, clinicopathologic characteristics, immune landscape, and therapeutic response were compared between these subtypes. The single-cell RNA sequencing (scRNA-seq) analysis and preliminary cell experiment were also conducted to verify our findings. RESULTS The altered bile acid profile and genetic variation of bile acid metabolism-related genes in cholangiocarcinoma were demonstrated. The cholangiocarcinoma was categorized into bile acid metabolism-active and -inactive subtypes with different prognoses, clinicopathologic characteristics, tumor microenvironments (TME) and therapeutic responses. This categorization was reproducible and predictable. Specifically, the bile acid metabolism-active subtype showed a poor prognosis with an immunosuppressive microenvironment and an inactive response to immunotherapy, while the bile acid metabolism-inactive subtype showed the opposite characteristics. Moreover, the scRNA-seq revealed that immunotherapy altered bile acid metabolism in TME of cholangiocarcinoma. Finally, a prognostic signature related to bile acid metabolism was developed, which exhibited strong power for prognostic assessment of cholangiocarcinoma. Consistently, these results were verified by immunohistochemistry, cell proliferation, migration, and apoptosis assays. CONCLUSION In conclusion, a novel cholangiocarcinoma classification based on bile acid metabolism was established. This classification was significant for the estimation of TME and prognosis.
Collapse
Affiliation(s)
- Mingxia Deng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jing Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Li Zhang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Yan Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
18
|
Jiang XY, Hu JJ, Wang R, Zhang WY, Jin QQ, Yang YT, Mei J, Hong L, Yao H, Tao F, Li JJ, Liu Y, Zhang L, Chen SX, Chen G, Song Y, Zhou SG. Cuproptosis-Associated lncRNA Gene Signature Establishes New Prognostic Profile and Predicts Immunotherapy Response in Endometrial Carcinoma. Biochem Genet 2024; 62:3439-3466. [PMID: 38108937 PMCID: PMC11427535 DOI: 10.1007/s10528-023-10574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/26/2023] [Indexed: 12/19/2023]
Abstract
Uterine corpus endometrial carcinoma (UCEC), a prevalent kind of cancerous tumor in female reproductive system that has a dismal prognosis in women worldwide. Given the very limited studies of cuproptosis-related lncRNAs (CRLs) in UCEC. Our purpose was to construct a prognostic profile based on CRLs and explore its assess prognostic value in UCEC victims and its correlation with the immunological microenvironment. METHODS 554 UCEC tumor samples and 23 normal samples' RNA-seq statistics and clinical details were compiled from data in the TCGA database. CRLs were obtained using Pearson correlation analysis. Using LASSO Cox regression, multivariate Cox regression, and univariate Cox regression analysis, six CRLs are confirmed to develop a risk prediction model at last.We identified two main molecular subtypes and observed that multilayer CRLs modifications were related to patient clinicopathological features, prognosis, and tumor microenvironment (TME) cell infiltration characteristics, and then we verified the prognostic hallmark of UCEC and examined its immunological landscape.Finally, using qRT-PCR, model hub genes' expression patterns were confirmed. RESULTS A unique CRL signature was established by the combination of six differently expressed CRLs that were highly linked with the prognosis of UCEC patients. According to their CRLs signatures, the patients were divided into two groups: the low-risk and the high-risk groups. Compared to individuals at high risk, patients at low risk had higher survival rates (p < 0.001). Additionally, Cox regression reveals that the profiles of lncRNAs linked to cuproptosis may independently predict prognosis in UCEC patients. The 1-, 3-, and 5-year risks' respective receiver operating characteristics (ROC) exhibited AUC values of 0.778, 0.810, and 0.854. Likewise, the signature could predict survival in different groups based on factors like stage, age, and grade, among others. Further investigation revealed differences between the different risk score groups in terms of drug sensitivity,immune cell infiltration,tumor mutation burden (TMB) score and microsatellite instability (MSI) score. Compared to the group of high risk, the low-risk group had greater rates of TMB and MSI. Results from qRT-PCR revealed that in UCEC vs normal tissues, AC026202.2, NRAV, AC079466.2, and AC090617.5 were upregulated,while LINC01545 and AL450384.1 were downregulated. CONCLUSIONS Our research clarified the relationship between CRLs signature and the immunological profile and prognosis of UCEC.This signature will establish the framework for future investigations into the endometrial cancer CRLs mechanism as well as the exploitation of new diagnostic tools and new therapeutic.
Collapse
Affiliation(s)
- Xi-Ya Jiang
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jing-Jing Hu
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Reproduction, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Rui Wang
- Department of Clinical Laboratory, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
| | - Wei-Yu Zhang
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Qin-Qin Jin
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yin-Ting Yang
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jie Mei
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Lin Hong
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hui Yao
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Feng Tao
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jie-Jie Li
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yu Liu
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Li Zhang
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shun-Xia Chen
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guo Chen
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yang Song
- Department of Pain, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Shu-Guang Zhou
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Anhui Province Maternity and Child Healthcare Hospital, Hefei, 230001, Anhui, China.
- Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
19
|
Huang Y, Li N, Jiang J, Pei Y, Gao S, Qian Y, Xing Y, Zhou T, Lian Y, Shi M. Metabolic reprogramming-related gene classifier distinguishes malignant from the benign pulmonary nodules. Heliyon 2024; 10:e37214. [PMID: 39296187 PMCID: PMC11409088 DOI: 10.1016/j.heliyon.2024.e37214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 07/02/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
The current existing classifiers for distinguishing malignant from benign pulmonary nodules is limited by effectiveness or clinical practicality. In our study, we aimed to develop and validate a gene classifier for lung cancer diagnosis. To identify the genes involved in this process, we used the weighted gene co-expression network analysis to analyze gene expression datasets from Gene Expression Omnibus (GEO). We identified the three most relevant modules associated with malignant nodules and performed functional enrichment analysis on them. The results indicated significant involvement in metabolic, immune-related, cell cycle, and viral-related processes. All three modules showed enrichment in metabolic reprogramming pathways. Based on these genes, we intersected genes from the three modules with metabolic reprogramming-related genes and further intersected with differentially expressed genes to get 78 genes. After machine learning algorithms and manual selection, we finally got a nine-gene classifier consisting of SEC24D, RPSA, PSME3, PSMD8, PSMB7, NCOA1, MED12, LPCAT1, and AKR1C3. Our developed and validated classifier-based model demonstrated good discrimination, with an area under the curve (AUC) of 0.763 in the development cohort, 0.744 in the internal validation cohort, and 0.718 in the external validation cohort, and outperformed previous clinical models. Moreover, the addition of nodule size improved the predictive capability of the classifier. We further verify the expression of the gene in the classifier using TCGA lung cancer samples and found eight of the genes showed significant differential expression in lung adenocarcinoma while all nine genes showed significant differential expression in lung squamous carcinoma. Our findings underscore the significance of metabolic reprogramming pathways in patients with malignant pulmonary nodules, and our gene classifier can assist clinicians in differentiating benign from malignant pulmonary nodules in clinical settings.
Collapse
Affiliation(s)
- Yongkang Huang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Na Li
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Soochow University, 9 Chongwen Road, Suzhou, 215004, Jiangsu, China
| | - Jie Jiang
- Department of Thoracic Surgery, the Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, 210003, Jiangsu, China
| | - Yongjian Pei
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Shiyuan Gao
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Yajuan Qian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Yufei Xing
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Tong Zhou
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Yixin Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Minhua Shi
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| |
Collapse
|
20
|
Jin Z, Wang X, Zhang X, Cheng S, Liu Y. Identification of two heterogeneous subtypes of hepatocellular carcinoma with distinct pathway activities and clinical outcomes based on gene set variation analysis. Front Genet 2024; 15:1441189. [PMID: 39323867 PMCID: PMC11423295 DOI: 10.3389/fgene.2024.1441189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Background High heterogeneity is an essential feature of malignant tumors. This study aims to reveal the drivers of hepatocellular carcinoma heterogeneity for prognostic stratification and to guide individualized treatment. Methods Omics data and clinical data for two HCC cohorts were derived from the Cancer Genome Atlas (TCGA) and the International Cancer Genome Atlas (ICGC), respectively. CNV data and methylation data were downloaded from the GSCA database. GSVA was used to estimate the transcriptional activity of KEGG pathways, and consensus clustering was used to categorize the HCC samples. The pRRophetic package was used to predict the sensitivity of samples to anticancer drugs. TIMER, MCPcounter, quanTIseq, and TIDE algorithms were used to assess the components of TME. LASSO and COX analyses were used to establish a prognostic gene signature. The biological role played by genes in HCC cells was confirmed by in vitro experiments. Results We classified HCC tissues into two categories based on the activity of prognostic pathways. Among them, the transcriptional profile of cluster A HCC is similar to that of normal tissue, dominated by cancer-suppressive metabolic pathways, and has a better prognosis. In contrast, cluster B HCC is dominated by high proliferative activity and has significant genetic heterogeneity. Meanwhile, cluster B HCC is often poorly differentiated, has a high rate of serum AFP positivity, is prone to microvascular invasion, and has shorter overall survival. In addition, we found that mutations, copy number variations, and aberrant methylation were also crucial drivers of the differences in heterogeneity between the two HCC subtypes. Meanwhile, the TME of the two HCC subtypes is also significantly different, which offers the possibility of precision immunotherapy for HCC patients. Finally, based on the prognostic value of molecular subtypes, we developed a gene signature that could accurately predict patients' OS. The riskscore quantified by the signature could evaluate the heterogeneity of HCC and guide clinical treatment. Finally, we confirmed through in vitro experiments that RFPL4B could promote the progression of Huh7 cells. Conclusion The molecular subtypes we identified effectively exposed the heterogeneity of HCC, which is important for discovering new effective therapeutic targets.
Collapse
Affiliation(s)
- Zhipeng Jin
- Department of Hepatopancreatobiliary Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Xin Wang
- Department of Hepatopancreatobiliary Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Xue Zhang
- Central Laboratory, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Siqi Cheng
- Department of Hepatopancreatobiliary Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Yefu Liu
- Department of Hepatopancreatobiliary Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Zhu C, Cheng Y, Yu Y, Zhang Y, Ren G. VIRMA promotes the progression of head and neck squamous cell carcinoma by regulating UBR5 mRNA and m6A levels. BIOMOLECULES & BIOMEDICINE 2024; 24:1244-1257. [PMID: 38577917 PMCID: PMC11379021 DOI: 10.17305/bb.2024.10358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/23/2024] [Accepted: 03/23/2024] [Indexed: 04/06/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a globally prevalent and lethal cancer form which precise mechanisms remain incompletely understood. Increasing evidence suggests that N6-methyladenosine (m6A) plays a crucial role in cancer progression. This study aimed to explore the biological function of m6A modification and vir-like m6A methyltransferase associated (VIRMA) in HNSCC. We conducted an analysis of VIRMA expression in HNSCC cells using The Cancer Genome Atlas (TCGA) database and employed reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting to assess its expression levels in HNSCC cell lines. Additionally, m6A levels in HNSCC cells were quantified, and the correlation between VIRMA expression levels and the clinical and pathological features of other genes was analyzed. Upon knocking down VIRMA levels, we assessed HNSCC cell proliferation, migration, and invasion and validated downstream genes using RT-qPCR and western blot. Our findings suggested that VIRMA, as an m6A-related regulator, may significantly influence HNSCC progression by regulating ubiquitin protein ligase E3 component N-recognin 5 (UBR5) through m6A modification. Therefore, VIRMA may serve as a prognostic biomarker.
Collapse
Affiliation(s)
- Chunyu Zhu
- Department of Oral and Maxillofacial Surgery, School of Stomatology and Stomatological Hospital, Hebei Medical University, Shijiazhuang, China
| | - Yameng Cheng
- The Key Laboratory of Oral Medicine in Hebei Province, School of Stomatology and Stomatological Hospital, Hebei Medical University, Shijiazhuang, China
| | - Yao Yu
- Hebei Provincial Clinical Research Center for Oral Diseases, Shijiazhuang, China
| | - Yanning Zhang
- The Key Laboratory of Oral Medicine in Hebei Province, School of Stomatology and Stomatological Hospital, Hebei Medical University, Shijiazhuang, China
| | - Guiyun Ren
- Department of Oral and Maxillofacial Surgery, School of Stomatology and Stomatological Hospital, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
22
|
Menegollo M, Bentham RB, Henriques T, Ng SQ, Ren Z, Esculier C, Agarwal S, Tong ETY, Lo C, Ilangovan S, Szabadkai Z, Suman M, Patani N, Ghanate A, Bryson K, Stein RC, Yuneva M, Szabadkai G. Multistate Gene Cluster Switches Determine the Adaptive Mitochondrial and Metabolic Landscape of Breast Cancer. Cancer Res 2024; 84:2911-2925. [PMID: 38924467 PMCID: PMC11372374 DOI: 10.1158/0008-5472.can-23-3172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/17/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Adaptive metabolic switches are proposed to underlie conversions between cellular states during normal development as well as in cancer evolution. Metabolic adaptations represent important therapeutic targets in tumors, highlighting the need to characterize the full spectrum, characteristics, and regulation of the metabolic switches. To investigate the hypothesis that metabolic switches associated with specific metabolic states can be recognized by locating large alternating gene expression patterns, we developed a method to identify interspersed gene sets by massive correlated biclustering and to predict their metabolic wiring. Testing the method on breast cancer transcriptome datasets revealed a series of gene sets with switch-like behavior that could be used to predict mitochondrial content, metabolic activity, and central carbon flux in tumors. The predictions were experimentally validated by bioenergetic profiling and metabolic flux analysis of 13C-labeled substrates. The metabolic switch positions also distinguished between cellular states, correlating with tumor pathology, prognosis, and chemosensitivity. The method is applicable to any large and heterogeneous transcriptome dataset to discover metabolic and associated pathophysiological states. Significance: A method for identifying the transcriptomic signatures of metabolic switches underlying divergent routes of cellular transformation stratifies breast cancer into metabolic subtypes, predicting their biology, architecture, and clinical outcome.
Collapse
Affiliation(s)
- Michela Menegollo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Robert B Bentham
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Tiago Henriques
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Seow Q Ng
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Clarinde Esculier
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Sia Agarwal
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Emily T Y Tong
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Clement Lo
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Sanjana Ilangovan
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Zorka Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Matteo Suman
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Neill Patani
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | | - Kevin Bryson
- Department of Computer Sciences, University College London, London, United Kingdom
| | - Robert C Stein
- Department of Oncology, University College London Hospitals, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
23
|
Neary B, Qiu P. Characterization of Expression-Based Gene Clusters Gives Insights into Variation in Patient Response to Cancer Therapies. Cancer Inform 2024; 23:11769351241271560. [PMID: 39238656 PMCID: PMC11375686 DOI: 10.1177/11769351241271560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/01/2024] [Indexed: 09/07/2024] Open
Abstract
Background Transcriptomics can reveal much about cellular activity, and cancer transcriptomics have been useful in investigating tumor cell behaviors. Patterns in transcriptome-wide gene expression can be used to investigate biological mechanisms and pathways that can explain the variability in patient response to cancer therapies. Methods We identified gene expression patterns related to patient drug response by clustering tumor gene expression data and selecting from the resulting gene clusters those where expression of cluster genes was related to patient survival on specific drugs. We then investigated these gene clusters for biological meaning using several approaches, including identifying common genomic locations and transcription factors whose targets were enriched in these clusters and performing survival analyses to support these candidate transcription factor-drug relationships. Results We identified gene clusters related to drug-specific survival, and through these, we were able to associate observed variations in patient drug response to specific known biological phenomena. Specifically, our analysis implicated 2 stem cell-related transcription factors, HOXB4 and SALL4, in poor response to temozolomide in brain cancers. In addition, expression of SNRNP70 and its targets were implicated in cetuximab response by 3 different analyses, although the mechanism remains unclear. We also found evidence that 2 cancer-related chromosomal structural changes may impact drug efficacy. Conclusion In this study, we present the gene clusters identified and the results of our systematic analysis linking drug efficacy to specific transcription factors, which are rich sources of potential mechanistic relationships impacting patient outcomes. We also highlight the most promising of these results, which were supported by multiple analyses and by previous research. We report these findings as promising avenues for independent validation and further research into cancer treatments and patient response.
Collapse
Affiliation(s)
- Bridget Neary
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| |
Collapse
|
24
|
Sarsenova M, Lawarde A, Pathare ADS, Saare M, Modhukur V, Soplepmann P, Terasmaa A, Käämbre T, Gemzell-Danielsson K, Lalitkumar PGL, Salumets A, Peters M. Endometriotic lesions exhibit distinct metabolic signature compared to paired eutopic endometrium at the single-cell level. Commun Biol 2024; 7:1026. [PMID: 39169201 PMCID: PMC11339455 DOI: 10.1038/s42003-024-06713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Current therapeutics of endometriosis focus on hormonal disruption of endometriotic lesions (ectopic endometrium, EcE). Recent findings show higher glycolysis utilization in EcE, suggesting non-hormonal strategy for disease treatment that addresses cellular metabolism. Identifying metabolically altered cell types in EcE is important for targeted metabolic drug therapy without affecting eutopic endometrium (EuE). Here, using single-cell RNA-sequencing, we examine twelve metabolic pathways in paired samples of EuE and EcE from women with confirmed endometriosis. We detect nine major cell types in both EuE and EcE. Metabolic pathways are most differentially regulated in perivascular, stromal, and endothelial cells, with the highest changes in AMPK signaling, HIF-1 signaling, glutathione metabolism, oxidative phosphorylation, and glycolysis. We identify transcriptomic co-activation of glycolytic and oxidative metabolism in perivascular and stromal cells of EcE, indicating a critical role of metabolic reprogramming in maintaining endometriotic lesion growth. Perivascular cells, involved in endometrial stroma repair and angiogenesis, may be potential targets for non-hormonal treatment of endometriosis.
Collapse
Affiliation(s)
- Meruert Sarsenova
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Ankita Lawarde
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Amruta D S Pathare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | | | - Anton Terasmaa
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Tuuli Käämbre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Kristina Gemzell-Danielsson
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Parameswaran Grace Luther Lalitkumar
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
- Competence Centre on Health Technologies, Tartu, Estonia.
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Maire Peters
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| |
Collapse
|
25
|
Li Y, Jiang LN, Zhao BK, Li ML, Jiang YY, Liu YS, Liu SH, Zhu L, Ye X, Zhao JM. Lecithin-cholesterol acyltransferase is a potential tumor suppressor and predictive marker for hepatocellular carcinoma metastasis. World J Gastrointest Oncol 2024; 16:3651-3671. [PMID: 39171187 PMCID: PMC11334038 DOI: 10.4251/wjgo.v16.i8.3651] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major cause of cancer mortality worldwide, and metastasis is the main cause of early recurrence and poor prognosis. However, the mechanism of metastasis remains poorly understood. AIM To determine the possible mechanism affecting HCC metastasis and provide a possible theoretical basis for HCC treatment. METHODS The candidate molecule lecithin-cholesterol acyltransferase (LCAT) was screened by gene microarray and bioinformatics analysis. The expression levels of LCAT in clinical cohort samples was detected by quantitative real-time polymerase chain reaction and western blotting. The proliferation, migration, invasion and tumor-forming ability were measured by Cell Counting Kit-8, Transwell cell migration, invasion, and clonal formation assays, respectively. Tumor formation was detected in nude mice after LCAT gene knockdown or overexpression. The immunohistochemistry for Ki67, E-cadherin, N-cadherin, matrix metalloproteinase 9 and vascular endothelial growth factor were performed in liver tissues to assess the effect of LCAT on HCC. Gene set enrichment analysis (GSEA) on various gene signatures were analyzed with GSEA version 3.0. Three machine-learning algorithms (random forest, support vector machine, and logistic regression) were applied to predict HCC metastasis in The Cancer Genome Atlas and GEO databases. RESULTS LCAT was identified as a novel gene relating to HCC metastasis by using gene microarray in HCC tissues. LCAT was significantly downregulated in HCC tissues, which is correlated with recurrence, metastasis and poor outcome of HCC patients. Functional analysis indicated that LCAT inhibited HCC cell proliferation, migration and invasion both in vitro and in vivo. Clinicopathological data showed that LCAT was negatively associated with HCC size and metastasis (HCC size ≤ 3 cm vs 3-9 cm, P < 0.001; 3-9 cm vs > 9 cm, P < 0.01; metastatic-free HCC vs extrahepatic metastatic HCC, P < 0.05). LCAT suppressed the growth, migration and invasion of HCC cell lines via PI3K/AKT/mTOR signaling. Our results indicated that the logistic regression model based on LCAT, TNM stage and the serum level of α-fetoprotein in HCC patients could effectively predict high metastatic risk HCC patients. CONCLUSION LCAT is downregulated at translational and protein levels in HCC and might inhibit tumor metastasis via attenuating PI3K/AKT/mTOR signaling. LCAT is a prognostic marker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Li-Na Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Bo-Kang Zhao
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Mei-Ling Li
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yi-Yun Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yi-Si Liu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Shu-Hong Liu
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Li Zhu
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xin Ye
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Jing-Min Zhao
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| |
Collapse
|
26
|
Chen H, Jing C, Shang L, Zhu X, Zhang R, Liu Y, Wang M, Xu K, Ma T, Jing H, Wang Z, Li X, Chong W, Li L. Molecular characterization and clinical relevance of metabolic signature subtypes in gastric cancer. Cell Rep 2024; 43:114424. [PMID: 38959111 DOI: 10.1016/j.celrep.2024.114424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/06/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024] Open
Abstract
Metabolic reprogramming dictates tumor molecular attributes and therapeutic potentials. However, the comprehensive metabolic characteristics in gastric cancer (GC) remain obscure. Here, metabolic signature-based clustering analysis identifies three subtypes with distinct molecular and clinical features: MSC1 showed better prognosis and upregulation of the tricarboxylic acid (TCA) cycle and lipid metabolism, combined with frequent TP53 and RHOA mutation; MSC2 had moderate prognosis and elevated nucleotide and amino acid metabolism, enriched by intestinal histology and mismatch repair deficient (dMMR); and MSC3 exhibited poor prognosis and enhanced glycan and energy metabolism, accompanied by diffuse histology and frequent CDH1 mutation. The Shandong Provincial Hospital (SDPH) in-house dataset with matched transcriptomic, metabolomic, and spatial-metabolomic analysis also validated these findings. Further, we constructed the metabolic subtype-related prognosis gene (MSPG) scoring model to quantify the activity of individual tumors and found a positive correlation with cuproptosis signaling. In conclusion, comprehensive recognition of the metabolite signature can enhance the understanding of diversity and heterogeneity in GC.
Collapse
Affiliation(s)
- Hao Chen
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Xingyu Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Ronghua Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Yuan Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Mingfei Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Kang Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Tianrong Ma
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Haiyan Jing
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Ze Wang
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xin Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China.
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan 250021, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China.
| |
Collapse
|
27
|
Köse SG, Güleç Taşkıran AE. Mechanisms of drug resistance in nutrient-depleted colorectal cancer cells: insights into lysosomal and mitochondrial drug sequestration. Biol Open 2024; 13:bio060448. [PMID: 39445740 PMCID: PMC11554266 DOI: 10.1242/bio.060448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
This Review delves into the mechanisms behind drug resistance in colorectal cancer (CRC), particularly examining the role of nutrient depletion and its contribution to multidrug resistance (MDR). The study highlights metabolic adaptations of cancer cells as well as metabolic adaptations of cancer cells under low nutrient availability, including shifts in glycolysis and lipid metabolism. It emphasizes the significance of MDR1 and its encoded efflux transporter, P-glycoprotein (P-gp/B1), in mediating drug resistance and how pathways such as HIF1α, AKT, and mTOR influence the expression of P-gp/B1 under limited nutrient availability. Additionally, the Review explores the dual roles of autophagy in drug sensitivity and resistance under nutrient limited conditions. It further investigates the involvement of lysosomes and mitochondria, focusing on their roles in drug sequestration and the challenges posed by lysosomal entrapment facilitated by non-enzymatic processes and ABC transporters like P-gp/B1. Finally, the Review underscores the importance of understanding the interplay between drug sequestration, lysosomal functions, nutrient depletion, and MDR1 gene modulation. It suggests innovative strategies, including structural modifications and nanotechnology, as promising approaches to overcoming drug resistance in cancer therapy.
Collapse
Affiliation(s)
- Serra Gülse Köse
- Molecular Biology and Genetics Department, Baskent University, Ankara 06790, Turkey
| | | |
Collapse
|
28
|
Huang XW, Li Y, Jiang LN, Zhao BK, Liu YS, Chen C, Zhao D, Zhang XL, Li ML, Jiang YY, Liu SH, Zhu L, Zhao JM. Nomogram for preoperative estimation of microvascular invasion risk in hepatocellular carcinoma. Transl Oncol 2024; 45:101986. [PMID: 38723299 PMCID: PMC11101742 DOI: 10.1016/j.tranon.2024.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/22/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024] Open
Abstract
Microvascular invasion (MVI) is an adverse prognostic indicator of tumor recurrence after surgery for hepatocellular carcinoma (HCC). Therefore, developing a nomogram for estimating the presence of MVI before liver resection is necessary. We retrospectively included 260 patients with pathologically confirmed HCC at the Fifth Medical Center of Chinese PLA General Hospital between January 2021 and April 2024. The patients were randomly divided into a training cohort (n = 182) for nomogram development, and a validation cohort (n = 78) to confirm the performance of the model (7:3 ratio). Significant clinical variables associated with MVI were then incorporated into the predictive nomogram using both univariate and multivariate logistic analyses. The predictive performance of the nomogram was assessed based on its discrimination, calibration, and clinical utility. Serum carnosine dipeptidase 1 ([CNDP1] OR 2.973; 95 % CI 1.167-7.575; p = 0.022), cirrhosis (OR 8.911; 95 % CI 1.922-41.318; p = 0.005), multiple tumors (OR 4.095; 95 % CI 1.374-12.205; p = 0.011), and tumor diameter ≥3 cm (OR 4.408; 95 % CI 1.780-10.919; p = 0.001) were independent predictors of MVI. Performance of the nomogram based on serum CNDP1, cirrhosis, number of tumors and tumor diameter was achieved with a concordance index of 0.833 (95 % CI 0.771-0.894) and 0.821 (95 % CI 0.720-0.922) in the training and validation cohorts, respectively. It fitted well in the calibration curves, and the decision curve analysis further confirmed its clinical usefulness. The nomogram, incorporating significant clinical variables and imaging features, successfully predicted the personalized risk of MVI in HCC preoperatively.
Collapse
Affiliation(s)
- Xiao-Wen Huang
- Medical School of Chinese PLA, Beijing, China; Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Li
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li-Na Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bo-Kang Zhao
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| | - Yi-Si Liu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chun Chen
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Dan Zhao
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xue-Li Zhang
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Mei-Ling Li
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yi-Yun Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shu-Hong Liu
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li Zhu
- Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing-Min Zhao
- Medical School of Chinese PLA, Beijing, China; Department of Pathology and Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
29
|
Zhang T, Zhao F, Hu Y, Wei J, Cui F, Lin Y, Jin Y, Sheng X. Environmental monobutyl phthalate exposure promotes liver cancer via reprogrammed cholesterol metabolism and activation of the IRE1α-XBP1s pathway. Oncogene 2024; 43:2355-2370. [PMID: 38879588 DOI: 10.1038/s41388-024-03086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/21/2024]
Abstract
Humans are widely exposed to phthalates, a major chemical plasticizer that accumulates in the liver. However, little is known about the impact of chronic phthalate exposure on liver cancer development. In this study, we applied a long-term cell culture model by treating the liver cancer cell HepG2 and normal hepatocyte L02 to environmental dosage of monobutyl phthalate (MBP), the main metabolite of phthalates. Interestingly, we found that long-term MBP exposure significantly accelerated the growth of HepG2 cells in vitro and in vivo, but barely altered the function of L02 cells. MBP exposure triggered reprogramming of lipid metabolism in HepG2 cells, where cholesterol accumulation subsequently activated the IRE1α-XBP1s axis of the unfolded protein response. As a result, the XBP1s-regulated gene sets and pathways contributed to the increased aggressiveness of HepG2 cells. In addition, we also showed that MBP-induced cholesterol accumulation fostered an immunosuppressive microenvironment by promoting tumor-associated macrophage polarization toward the M2 type. Together, these results suggest that environmental phthalates exposure may facilitate liver cancer progression, and alerts phthalates exposure to patients who already harbor liver tumors.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
- School of Environmental Science and Engineering, Hainan University, Haikou, 570228, China
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Faming Zhao
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
- School of Environmental Science and Engineering, Hainan University, Haikou, 570228, China
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanxia Hu
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
- School of Environmental Science and Engineering, Hainan University, Haikou, 570228, China
| | - Jinlan Wei
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengzhen Cui
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Yahang Lin
- Department of Neurology, Wuhan Fourth Hospital, Wuhan, 430033, China
| | - Yang Jin
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway
| | - Xia Sheng
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China.
- School of Environmental Science and Engineering, Hainan University, Haikou, 570228, China.
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
30
|
Pan JJ, Xie SZ, Zheng X, Xu JF, Xu H, Yin RQ, Luo YL, Shen L, Chen ZR, Chen YR, Yu SZ, Lu L, Zhu WW, Lu M, Qin LX. Acetyl-CoA metabolic accumulation promotes hepatocellular carcinoma metastasis via enhancing CXCL1-dependent infiltration of tumor-associated neutrophils. Cancer Lett 2024; 592:216903. [PMID: 38670307 DOI: 10.1016/j.canlet.2024.216903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
High levels of acetyl-CoA are considered a key metabolic feature of metastatic cancers. However, the impacts of acetyl-CoA metabolic accumulation on cancer microenvironment remodeling are poorly understood. In this study, using human hepatocellular carcinoma (HCC) tissues and orthotopic xenograft models, we found a close association between high acetyl-CoA levels in HCCs, increased infiltration of tumor-associated neutrophils (TANs) in the cancer microenvironment and HCC metastasis. Cytokine microarray and enzyme-linked immunosorbent assays (ELISA) revealed the crucial role of the chemokine (C-X-C motif) ligand 1(CXCL1). Mechanistically, acetyl-CoA accumulation induces H3 acetylation-dependent upregulation of CXCL1 gene expression. CXCL1 recruits TANs, leads to neutrophil extracellular traps (NETs) formation and promotes HCC metastasis. Collectively, our work linked the accumulation of acetyl-CoA in HCC cells and TANs infiltration, and revealed that the CXCL1-CXC receptor 2 (CXCR2)-TANs-NETs axis is a potential target for HCCs with high acetyl-CoA levels.
Collapse
Affiliation(s)
- Jun-Jie Pan
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Sun-Zhe Xie
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Xin Zheng
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Jian-Feng Xu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Hao Xu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Rui-Qi Yin
- Department of Infectious Diseases, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Yun-Ling Luo
- Department of Infectious Diseases, Rui'an People's Hospital, Wenzhou Medical University, 168 Ruifeng Avenue, Zhejiang 325200, China
| | - Li Shen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zheng-Ru Chen
- Department of Infectious Diseases, Rui'an People's Hospital, Wenzhou Medical University, 168 Ruifeng Avenue, Zhejiang 325200, China
| | - Yi-Ran Chen
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Shi-Zhe Yu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Lu Lu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Wen-Wei Zhu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China.
| | - Ming Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Lun-Xiu Qin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, 130 Dongan Road, Shanghai 200032, China.
| |
Collapse
|
31
|
Chaubal R, Gardi N, Joshi S, Pantvaidya G, Kadam R, Vanmali V, Hawaldar R, Talker E, Chitra J, Gera P, Bhatia D, Kalkar P, Gurav M, Shetty O, Desai S, Krishnan NM, Nair N, Parmar V, Dutt A, Panda B, Gupta S, Badwe R. Surgical Tumor Resection Deregulates Hallmarks of Cancer in Resected Tissue and the Surrounding Microenvironment. Mol Cancer Res 2024; 22:572-584. [PMID: 38394149 PMCID: PMC11148542 DOI: 10.1158/1541-7786.mcr-23-0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/24/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
UNLABELLED Surgery exposes tumor tissue to severe hypoxia and mechanical stress leading to rapid gene expression changes in the tumor and its microenvironment, which remain poorly characterized. We biopsied tumor and adjacent normal tissues from patients with breast (n = 81) and head/neck squamous cancers (HNSC; n = 10) at the beginning (A), during (B), and end of surgery (C). Tumor/normal RNA from 46/81 patients with breast cancer was subjected to mRNA-Seq using Illumina short-read technology, and from nine patients with HNSC to whole-transcriptome microarray with Illumina BeadArray. Pathways and genes involved in 7 of 10 known cancer hallmarks, namely, tumor-promoting inflammation (TNF-A, NFK-B, IL18 pathways), activation of invasion and migration (various extracellular matrix-related pathways, cell migration), sustained proliferative signaling (K-Ras Signaling), evasion of growth suppressors (P53 signaling, regulation of cell death), deregulating cellular energetics (response to lipid, secreted factors, and adipogenesis), inducing angiogenesis (hypoxia signaling, myogenesis), and avoiding immune destruction (CTLA4 and PDL1) were significantly deregulated during surgical resection (time points A vs. B vs. C). These findings were validated using NanoString assays in independent pre/intra/post-operative breast cancer samples from 48 patients. In a comparison of gene expression data from biopsy (analogous to time point A) with surgical resection samples (analogous to time point C) from The Cancer Genome Atlas study, the top deregulated genes were the same as identified in our analysis, in five of the seven studied cancer types. This study suggests that surgical extirpation deregulates the hallmarks of cancer in primary tumors and adjacent normal tissue across different cancers. IMPLICATIONS Surgery deregulates hallmarks of cancer in human tissue.
Collapse
Affiliation(s)
- Rohan Chaubal
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Nilesh Gardi
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Shalaka Joshi
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Gouri Pantvaidya
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Rasika Kadam
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Vaibhav Vanmali
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Clinical Research Secretariat, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Rohini Hawaldar
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Clinical Research Secretariat, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Elizabeth Talker
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Jaya Chitra
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Poonam Gera
- Biorepository, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Dimple Bhatia
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Prajakta Kalkar
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Mamta Gurav
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Omshree Shetty
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Sangeeta Desai
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | | | - Nita Nair
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Vani Parmar
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- 3D Printing Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Amit Dutt
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Binay Panda
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sudeep Gupta
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Rajendra Badwe
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| |
Collapse
|
32
|
Xu Z, He D, Huang L, Deng K, Jiang W, Qin J, Zheng Z, Zheng T, Li S. Metabolic reprogramming-driven homologous recombination and TCA cycle dysregulation contribute to poor prognoses in lung adenocarcinoma. J Cell Mol Med 2024; 28:e18406. [PMID: 38822457 PMCID: PMC11142899 DOI: 10.1111/jcmm.18406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/04/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024] Open
Abstract
Increasing evidence has shown that homologous recombination (HR) and metabolic reprogramming are essential for cellular homeostasis. These two processes are independent as well as closely intertwined. Nevertheless, they have rarely been reported in lung adenocarcinoma (LUAD). We analysed the genomic, immune microenvironment and metabolic microenvironment features under different HR activity states. Using cell cycle, EDU and cell invasion assays, we determined the impacts of si-SHFM1 on the LUAD cell cycle, proliferation and invasion. The levels of isocitrate dehydrogenase (IDH) and α-ketoglutarate dehydrogenase (α-KGDH) were determined by ELISA in the NC and si-SHFM1 groups of A549 cells. Finally, cell samples were used to extract metabolites for HPIC-MS/MS to analyse central carbon metabolism. We found that high HR activity was associated with a poor prognosis in LUAD, and HR was an independent prognostic factor for TCGA-LUAD patients. Moreover, LUAD samples with a high HR activity presented low immune infiltration levels, a high degree of genomic instability, a good response status to immune checkpoint blockade therapy and a high degree of drug sensitivity. The si-SHFM1 group presented a significantly higher proportion of cells in the G0/G1 phase, lower levels of DNA replication, and significantly lower levels of cell migration and both TCA enzymes. Our current results indicated that there is a strong correlation between HR and the TCA cycle in LUAD. The TCA cycle can promote SHFM1-mediated HR in LUAD, raising their activities, which can finally result in a poor prognosis and impair immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Zhanyu Xu
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Dongming He
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Liuliu Huang
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Kun Deng
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wei Jiang
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Junqi Qin
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Zhiwen Zheng
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Tiaozhan Zheng
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Shikang Li
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
33
|
Wang W, Li C, Dai Y, Wu Q, Yu W. Unraveling metabolic characteristics and clinical implications in gastric cancer through single-cell resolution analysis. Front Mol Biosci 2024; 11:1399679. [PMID: 38831933 PMCID: PMC11145399 DOI: 10.3389/fmolb.2024.1399679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024] Open
Abstract
Background: Gastric cancer is a highly prevalent malignant neoplasm. Metabolic reprogramming is intricately linked to both tumorigenesis and cancer immune evasion. The advent of single-cell RNA sequencing technology provides a novel perspective for evaluating cellular metabolism. This study aims to comprehensively investigate the metabolic pathways of various cell types in tumor and normal samples at high resolution and delve into the intricate regulatory mechanisms governing the metabolic activity of malignant cells in gastric cancer. Methods: Utilizing single-cell RNA sequencing data from gastric cancer, we constructed metabolic landscape maps for different cell types in tumor and normal samples. Employing unsupervised clustering, we categorized malignant cells in tumor samples into high and low metabolic subclusters and further explored the characteristics of these subclusters. Results: Our research findings indicate that epithelial cells in tumor samples exhibit significantly higher activity in most KEGG metabolic pathways compared to other cell types. Unsupervised clustering, based on the scores of metabolic pathways, classified malignant cells into high and low metabolic subclusters. In the high metabolic subcluster, it demonstrated the potential to induce a stronger immune response, correlating with a relatively favorable prognosis. In the low metabolic subcluster, a subset of cells resembling cancer stem cells (CSCs) was identified, and its prognosis was less favorable. Furthermore, a set of risk genes associated with this subcluster was discovered. Conclusion: This study reveals the intricate regulatory mechanisms governing the metabolic activity of malignant cells in gastric cancer, offering new perspectives for improving prognosis and treatment strategies.
Collapse
Affiliation(s)
- Wenyue Wang
- School of Life Sciences, Tianjin University, Tianjin, China
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, China
| | - Conghui Li
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yuting Dai
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qingfa Wu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Weiqiang Yu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, China
| |
Collapse
|
34
|
Tang J, Liu Y, Wang Y, Zhang Z, Nie J, Wang X, Ai S, Li J, Gao Y, Li C, Cheng C, Su S, Chen S, Zhang P, Lu R. Deciphering metabolic heterogeneity in retinoblastoma unravels the role of monocarboxylate transporter 1 in tumor progression. Biomark Res 2024; 12:48. [PMID: 38730450 PMCID: PMC11088057 DOI: 10.1186/s40364-024-00596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Tumors exhibit metabolic heterogeneity, influencing cancer progression. However, understanding metabolic diversity in retinoblastoma (RB), the primary intraocular malignancy in children, remains limited. METHODS The metabolic landscape of RB was constructed based on single-cell transcriptomic sequencing from 11 RB and 5 retina samples. Various analyses were conducted, including assessing overall metabolic activity, metabolic heterogeneity, and the correlation between hypoxia and metabolic pathways. Additionally, the expression pattern of the monocarboxylate transporter (MCT) family in different cell clusters was examined. Validation assays of MCT1 expression and function in RB cell lines were performed. The therapeutic potential of targeting MCT1 was evaluated using an orthotopic xenograft model. A cohort of 47 RB patients was analyzed to evaluate the relationship between MCT1 expression and tumor invasion. RESULTS Distinct metabolic patterns in RB cells, notably increased glycolysis, were identified. This metabolic heterogeneity correlated closely with hypoxia. MCT1 emerged as the primary monocarboxylate transporter in RB cells. Disrupting MCT1 altered cell viability and energy metabolism. In vivo studies using the MCT1 inhibitor AZD3965 effectively suppressed RB tumor growth. Additionally, a correlation between MCT1 expression and optic nerve invasion in RB samples suggested prognostic implications. CONCLUSIONS This study enhances our understanding of RB metabolic characteristics at the single-cell level, highlighting the significance of MCT1 in RB pathogenesis. Targeting MCT1 holds promise as a therapeutic strategy for combating RB, with potential prognostic implications.
Collapse
Affiliation(s)
- Junjie Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yaoming Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yinghao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zhihui Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jiahe Nie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyue Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Siming Ai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jinmiao Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yang Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Cheng Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Chao Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Shicai Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Shuxia Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Rong Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
35
|
Tahmori H, Ghahremani H, Nabati S, Mehdikhani F, Mirlohi M, Salami S, Sirati-Sabet M. Epithelial-mesenchymal transition-related signaling pathways in gastric Cancer cells distinctively respond to long-term experimental ketosis. Mol Biol Rep 2024; 51:641. [PMID: 38727798 DOI: 10.1007/s11033-024-09571-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 04/17/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND The interrelationship between cellular metabolism and the epithelial-to-mesenchymal transition (EMT) process has made it an interesting topic to investigate the adjuvant effect of therapeutic diets in the treatment of cancers. However, the findings are controversial. In this study, the effects of glucose limitation along and with the addition of beta-hydroxybutyrate (bHB) were examined on the expression of specific genes and proteins of EMT, Wnt, Hedgehog, and Hippo signaling pathways, and also on cellular behavior of gastric cancer stem-like (MKN-45) and non-stem-like (KATO III) cells. METHODS AND RESULTS The expression levels of chosen genes and proteins studied in cancer cells gradually adopted a low-glucose condition of one-fourth, along and with the addition of bHB, and compared to the unconditioned control cells. The long-term switching of the metabolic fuels successfully altered the expression profiles and behaviors of both gastric cancer cells. However, the results for some changes were the opposite. Glucose limitation along and with the addition of bHB reduced the CD44+ population in MKN-45 cells. In KATO III cells, glucose restriction increased the CD44+ population. Glucose deprivation alleviated EMT-related signaling pathways in MKN-45 cells but stimulated EMT in KATO III cells. Interestingly, bHB enrichment reduced the beneficial effect of glucose starvation in MKN-45 cells, but also alleviated the adverse effects of glucose restriction in KATO III cells. CONCLUSIONS The findings of this research clearly showed that some controversial results in clinical trials for ketogenic diet in cancer patients stemmed from the different signaling responses of various cells to the metabolic changes in a heterogeneous cancer mass.
Collapse
Affiliation(s)
- Haniyeh Tahmori
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Hossein Ghahremani
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Saeedeh Nabati
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Fatemeh Mehdikhani
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Maryamsadat Mirlohi
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Siamak Salami
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran
| | - Majid Sirati-Sabet
- Shahid Beheshti University of Medical Sciences School of Medicine, Tehran, Iran.
| |
Collapse
|
36
|
Wang K, Zerdes I, Johansson HJ, Sarhan D, Sun Y, Kanellis DC, Sifakis EG, Mezheyeuski A, Liu X, Loman N, Hedenfalk I, Bergh J, Bartek J, Hatschek T, Lehtiö J, Matikas A, Foukakis T. Longitudinal molecular profiling elucidates immunometabolism dynamics in breast cancer. Nat Commun 2024; 15:3837. [PMID: 38714665 PMCID: PMC11076527 DOI: 10.1038/s41467-024-47932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/12/2024] [Indexed: 05/10/2024] Open
Abstract
Although metabolic reprogramming within tumor cells and tumor microenvironment (TME) is well described in breast cancer, little is known about how the interplay of immune state and cancer metabolism evolves during treatment. Here, we characterize the immunometabolic profiles of tumor tissue samples longitudinally collected from individuals with breast cancer before, during and after neoadjuvant chemotherapy (NAC) using proteomics, genomics and histopathology. We show that the pre-, on-treatment and dynamic changes of the immune state, tumor metabolic proteins and tumor cell gene expression profiling-based metabolic phenotype are associated with treatment response. Single-cell/nucleus RNA sequencing revealed distinct tumor and immune cell states in metabolism between cold and hot tumors. Potential drivers of NAC based on above analyses were validated in vitro. In summary, the study shows that the interaction of tumor-intrinsic metabolic states and TME is associated with treatment outcome, supporting the concept of targeting tumor metabolism for immunoregulation.
Collapse
Affiliation(s)
- Kang Wang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Henrik J Johansson
- Department of Oncology-Pathology, Karolinska Institutet, and Science for Life Laboratory, Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yizhe Sun
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Dimitris C Kanellis
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Xingrong Liu
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Loman
- Department of Hematology, Oncology and Radiation Physics, Lund University Hospital, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Jiri Bartek
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Danish Cancer Institute, DK-2100, Copenhagen, Denmark
| | - Thomas Hatschek
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Karolinska Institutet, and Science for Life Laboratory, Stockholm, Sweden
- Division of Pathology, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden.
| |
Collapse
|
37
|
Berrell N, Sadeghirad H, Blick T, Bidgood C, Leggatt GR, O'Byrne K, Kulasinghe A. Metabolomics at the tumor microenvironment interface: Decoding cellular conversations. Med Res Rev 2024; 44:1121-1146. [PMID: 38146814 DOI: 10.1002/med.22010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023]
Abstract
Cancer heterogeneity remains a significant challenge for effective cancer treatments. Altered energetics is one of the hallmarks of cancer and influences tumor growth and drug resistance. Studies have shown that heterogeneity exists within the metabolic profile of tumors, and personalized-combination therapy with relevant metabolic interventions could improve patient response. Metabolomic studies are identifying novel biomarkers and therapeutic targets that have improved treatment response. The spatial location of elements in the tumor microenvironment are becoming increasingly important for understanding disease progression. The evolution of spatial metabolomics analysis now allows scientists to deeply understand how metabolite distribution contributes to cancer biology. Recently, these techniques have spatially resolved metabolite distribution to a subcellular level. It has been proposed that metabolite mapping could improve patient outcomes by improving precision medicine, enabling earlier diagnosis and intraoperatively identifying tumor margins. This review will discuss how altered metabolic pathways contribute to cancer progression and drug resistance and will explore the current capabilities of spatial metabolomics technologies and how these could be integrated into clinical practice to improve patient outcomes.
Collapse
Affiliation(s)
- Naomi Berrell
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Habib Sadeghirad
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tony Blick
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Charles Bidgood
- APCRC-Q, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Graham R Leggatt
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Ken O'Byrne
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
38
|
Sun J, Wang Y, Zhang K, Shi S, Gao X, Jia X, Cong B, Zheng C. Molecular subtype construction and prognosis model for stomach adenocarcinoma characterized by metabolism-related genes. Heliyon 2024; 10:e28413. [PMID: 38596054 PMCID: PMC11002599 DOI: 10.1016/j.heliyon.2024.e28413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Background Metabolic reprogramming is implicated in cancer progression. However, the impact of metabolism-associated genes in stomach adenocarcinomas (STAD) has not been thoroughly reviewed. Herein, we characterized metabolic transcription-correlated STAD subtypes and evaluated a metabolic RiskScore for evaluation survival. Method Genes related to metabolism were gathered from previous study and metabolic subtypes were screened using ConsensusClusterPlus in TCGA-STAD and GSE66229 dataset. The ssGSEA, MCP-Count, ESTIMATE and CIBERSORT determined the immune infiltration. A RiskScore model was established using the WGCNA and LASSO Cox regression in the TCGA-STAD queue and verified in the GSE66229 datasets. RT-qPCR was employed to measure the mRNA expressions of genes in the model. Result Two metabolism-related subtypes (C1 and C2) of STAD were constructed on account of the expression profiles of 113 prognostic metabolism genes with different immune outcomes and apparently distinct metabolic characteristic. The overall survival (OS) of C2 subtype was shorter than that of C1 subtype. Four metabolism-associated genes in turquoise model, which closely associated with C2 subtype, were employed to build the RiskScore (MATN3, OSBPL1A, SERPINE1, CPNE8) in TCGA-train dataset. Patients developed a poorer prognosis if they had a high RiskScore than having a low RiskScore. The promising effect of RiskScore was verified in the TCGA-test, TCGA-STAD and GSE66229 datasets. The prediction reliability of the RiskScore was validated by time-dependent receiver operating characteristic curve (ROC) and nomogram. Moreover, samples with high RiskScore had an enhanced immune status and TIDE score. Moreover, MATN3, OSBPL1A, SERPINE1 and CPNE8 mRNA levels were all elevated in SGC7901 cells. Inhibition of OSBPL1A decreased SGC7901 cells invasion numbers. Conclusion This work provided a new perspective into heterogeneity in metabolism and its association with immune escape in STAD. RiskScore was considered to be a strong prognostic label that could help individualize the treatment of STAD patients.
Collapse
Affiliation(s)
- Jie Sun
- Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Yuanyuan Wang
- Department of Oncology and Hematology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| | - Kai Zhang
- General Surgery Department, Wenshang County People's Hospital, Wenshang, 272501, China
| | - Sijia Shi
- Shandong Provincial Hospital, Jinan, 250001, China
| | - Xinxin Gao
- Gastrointestinal Surgery, Shandong First Medical University Affiliated Provincial Hospital, Jinan, 250001, China
| | - Xianghao Jia
- Gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, 250001, China
| | - Bicong Cong
- Gastrointestinal Surgery, Shandong First Medical University Affiliated Provincial Hospital, Jinan, 250001, China
| | - Chunning Zheng
- Gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, 250001, China
| |
Collapse
|
39
|
Xu YJ, Huo YC, Zhao QT, Liu JY, Tian YJ, Yang LL, Zhang Y. NOX4 promotes tumor progression through the MAPK-MEK1/2-ERK1/2 axis in colorectal cancer. World J Gastrointest Oncol 2024; 16:1421-1436. [PMID: 38660653 PMCID: PMC11037073 DOI: 10.4251/wjgo.v16.i4.1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/04/2024] [Accepted: 02/07/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Metabolic reprogramming plays a key role in cancer progression and clinical outcomes; however, the patterns and primary regulators of metabolic reprogramming in colorectal cancer (CRC) are not well understood. AIM To explore the role of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) in promoting progression of CRC. METHODS We evaluated the expression and function of dysregulated and survival-related metabolic genes using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. Consensus clustering was used to cluster CRC based on dysregulated metabolic genes. A prediction model was constructed based on survival-related metabolic genes. Sphere formation, migration, invasion, proliferation, apoptosis and clone formation was used to evaluate the biological function of NOX4 in CRC. mRNA sequencing was utilized to explore the alterations of gene expression NOX4 over-expression tumor cells. In vivo subcutaneous and lung metastasis mouse tumor model was used to explore the effect of NOX4 on tumor growth. RESULTS We comprehensively analyzed 3341 metabolic genes in CRC and identified three clusters based on dysregulated metabolic genes. Among these genes, NOX4 was highly expressed in tumor tissues and correlated with worse survival. In vitro, NOX4 overexpression induced clone formation, migration, invasion, and stemness in CRC cells. Furthermore, RNA-sequencing analysis revealed that NOX4 overexpression activated the mitogen-activated protein kinase-MEK1/2-ERK1/2 signaling pathway. Trametinib, a MEK1/2 inhibitor, abolished the NOX4-mediated tumor progression. In vivo, NOX4 overexpression promoted subcutaneous tumor growth and lung metastasis, whereas trametinib treatment can reversed the metastasis. CONCLUSION Our study comprehensively analyzed metabolic gene expression and highlighted the importance of NOX4 in promoting CRC metastasis, suggesting that trametinib could be a potential therapeutic drugs of CRC clinical therapy targeting NOX4.
Collapse
Affiliation(s)
- Yu-Jie Xu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Department of Oncology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou 450003, Henan Province, China
| | - Ya-Chang Huo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Qi-Tai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jin-Yan Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yi-Jun Tian
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Lei-Lei Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
40
|
Lei J, Luo J, Liu Q, Wang X. Identifying cancer subtypes based on embryonic and hematopoietic stem cell signatures in pan-cancer. Cell Oncol (Dordr) 2024; 47:587-605. [PMID: 37821797 DOI: 10.1007/s13402-023-00886-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
PURPOSE Cancer cells with stem cell-like properties may contribute to cancer development and therapy resistance. The advancement of multi-omics technology has sparked interest in exploring cancer stemness from a multi-omics perspective. However, there is a limited number of studies that have attempted to subtype cancer by combining different types of stem cell signatures. METHODS In this study, 10,323 cancer specimens from 33 TCGA cancer types were clustered based on the enrichment scores of six stemness gene sets, representing two types of stem cell backgrounds: embryonic stem cells (ESCs) and hematopoietic stem cells (HSCs). RESULTS We identified four subtypes of pan-cancer, termed StC1, StC2, StC3 and StC4, which displayed distinct molecular and clinical features, including stemness, genome integrity, intratumor heterogeneity, methylation levels, tumor microenvironment, tumor progression, responses to chemotherapy and immunotherapy, and survival prognosis. Importantly, this subtyping method for pan-cancer is reproducible at the protein level. CONCLUSION Our findings indicate that the ESC signature is an adverse prognostic factor in cancer, while the HSC signature and ratio of HSC/ESC signatures are positive prognostic factors. The subtyping of cancer based on ESC and HSC signatures may provide insights into cancer biology and clinical implications of cancer.
Collapse
Affiliation(s)
- Jiali Lei
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiangti Luo
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Qian Liu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
41
|
Wang Z, Zhang Y, Yang X, Zhang T, Li Z, Zhong Y, Fang Y, Chong W, Chen H, Lu M. Genetic and molecular characterization of metabolic pathway-based clusters in esophageal squamous cell carcinoma. Sci Rep 2024; 14:6200. [PMID: 38486026 PMCID: PMC10940668 DOI: 10.1038/s41598-024-56391-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive types of squamous cell carcinoma and represents a significant proportion of esophageal cancer. Metabolic reprogramming plays a key role in the occurrence and development of ESCC. Unsupervised clustering analysis was employed to stratify ESCC samples into three clusters: MPC1-lipid type, MPC2-amino acid type, and MPC3-energy type, based on the enrichment scores of metabolic pathways extracted from the Reactome database. The MPC3 cluster exhibited characteristics of energy metabolism, with heightened glycolysis, cofactors, and nucleotide metabolism, showing a trend toward increased aggressiveness and poorer survival rates. On the other hand, MPC1 and MPC2 primarily involved lipid and amino acid metabolism, respectively. In addition, liquid chromatography‒mass spectrometry-based metabolite profiles and potential therapeutic agents were explored and compared among ESCC cell lines with different MPCs. MPC3 amplified energy metabolism markers, especially carnitines. In contrast, MPC1 and MPC2 predominantly had elevated levels of lipids (primarily triacylglycerol) and amino acids, respectively. Furthermore, MPC3 demonstrated a suboptimal clinical response to PD-L1 immunotherapy but showed increased sensitivity to the doramapimod chemotherapy regimen, as evident from drug sensitivity evaluations. These insights pave the way for a more personalized therapeutic approach, potentially enhancing treatment precision for ESCC patients.
Collapse
Affiliation(s)
- Ze Wang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Zhang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Tongchao Zhang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhen Li
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yang Zhong
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Fang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Ming Lu
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
42
|
Weistuch C, Murgas KA, Zhu J, Norton L, Dill KA, Tannenbaum AR, Deasy JO. Functional transcriptional signatures for tumor-type-agnostic phenotype prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.12.536595. [PMID: 37090606 PMCID: PMC10120658 DOI: 10.1101/2023.04.12.536595] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Cancer transcriptional patterns exhibit both shared and unique features across diverse cancer types, but whether these patterns are sufficient to characterize the full breadth of tumor phenotype heterogeneity remains an open question. We hypothesized that cancer transcriptional diversity mirrors patterns in normal tissues optimized for distinct functional tasks. Starting with normal tissue transcriptomic profiles, we use non-negative matrix factorization to derive six distinct transcriptomic phenotypes, called archetypes, which combine to describe both normal tissue patterns and variations across a broad spectrum of malignancies. We show that differential enrichment of these signatures correlates with key tumor characteristics, including overall patient survival and drug sensitivity, independent of clinically actionable DNA alterations. Additionally, we show that in HR+/HER2- breast cancers, metastatic tumors adopt transcriptomic signatures consistent with the invaded tissue. Broadly, our findings suggest that cancer often arrogates normal tissue transcriptomic characteristics as a component of both malignant progression and drug response. This quantitative framework provides a strategy for connecting the diversity of cancer phenotypes and could potentially help manage individual patients.
Collapse
Affiliation(s)
- Corey Weistuch
- Memorial Sloan Kettering Cancer Center, Department of Medical
Physics
| | - Kevin A. Murgas
- Stony Brook University, Department of Biomedical
Informatics
| | - Jiening Zhu
- Stony Brook University, Department of Applied Mathematics and
Statistics
| | - Larry Norton
- Memorial Sloan Kettering Cancer Center, Department of
Medicine
| | - Ken A. Dill
- Stony Brook University, Laufer Center for Physical and
Quantitative Biology
| | - Allen R. Tannenbaum
- Stony Brook University, Department of Applied Mathematics and
Statistics
- Stony Brook University, Department of Computer Science
| | - Joseph O. Deasy
- Memorial Sloan Kettering Cancer Center, Department of Medical
Physics
| |
Collapse
|
43
|
Zhang X, Li W, Liu T, Guo H, Sun Q, Li B. Heterogeneity of Lipid Metabolism and its Clinical and Immune Correlation in Lung Adenocarcinoma. Curr Med Chem 2024; 31:1561-1577. [PMID: 37594166 DOI: 10.2174/0929867331666230818144416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION The role of lipid metabolism in lung adenocarcinoma (LUAD) is not completely researched. Lipid metabolism reprogramming is a characteristic of malignancies and contributes to carcinogenesis and progression. The transcriptome and scRNA- seq data and clinical information were downloaded from the public databases. METHODS Lipid metabolism pathways were collected from the MSigDB database, and molecular subtypes were classified based on lipid metabolism features via consensus clustering. The bidirectional crosstalk between immune cells and malignant cells was analyzed. Differences in lipid metabolism at the single-cell level and their correlation with the tumor microenvironment (TME) were also studied. LUAD patients were classified into two subtypes, showing distinct mutation and lipid metabolism features based on lipid metabolism characteristics. Meanwhile, significant differences in the overall survival, clinical characteristics, and immune landscape were observed between the two subtypes. We also found that clust1 had higher oxidative stress status. There were 116 differentially expressed genes between the two subtypes, which were significantly associated with cell cycle progression. We identified 4001 immune cells, including 483 malignant cells and 3518 normal cells, and found active intercellular communication and significant differences in lipid metabolism characteristics between the malignant cells and normal cells. Furthermore, several lipid metabolism pathways were found to be associated with TME factors, including hypoxia and angiogenesis. RESULT The current findings indicated that lipid metabolism was involved in the development and cellular heterogeneity of LUAD and revealed widespread reprogramming across multiple cellular elements in the TME of LUAD. CONCLUSION This characterization improved the current understanding of tumor biology and enabled the identification of novel targets for immunotherapy.
Collapse
Affiliation(s)
- Xugang Zhang
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| | - Weiqing Li
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| | - Taorui Liu
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| | - Huiqin Guo
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| | - Qianqian Sun
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| | - Baozhong Li
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100000, China
| |
Collapse
|
44
|
He B, Hu Y, Chen H, Xie X, Gong C, Li Z, Chen Y, Xiao Y, Yang S. Modification patterns and metabolic characteristics of m 6A regulators in digestive tract tumors. Heliyon 2024; 10:e24235. [PMID: 38298699 PMCID: PMC10828661 DOI: 10.1016/j.heliyon.2024.e24235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/29/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
M6A is essential for tumor occurrence and progression. The expression patterns of m6A regulators differ in various kinds of tumors. Transcriptomic expression statistics together with clinical data from a database were analyzed to distinguish patients with digestive tract tumors. Based on the expression patterns of diverse m6A regulators, patients were divided into several clusters. Survival analysis suggested significant differences in patient prognosis among the m6A clusters. The results showed overlapping of m6A expression patterns with energy metabolism and nucleotide metabolism. Functional analyses imply that m6A modifications in tumor cells probably drive metabolic reprogramming to sustain rapid proliferation of cancer cells. Our analysis highlights the m6A risk characterizes various kinds of metabolic features and predicts chemotherapy sensitivity in digestive tract tumors, providing evidence for m6A regulators as markers to predict patient outcomes.
Collapse
Affiliation(s)
| | | | - Hui Chen
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Xia Xie
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Chunli Gong
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Zhibin Li
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yang Chen
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yufeng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| |
Collapse
|
45
|
Wu C, Tan J, Shen H, Deng C, Kleber C, Osterhoff G, Schopow N. Exploring the relationship between metabolism and immune microenvironment in osteosarcoma based on metabolic pathways. J Biomed Sci 2024; 31:4. [PMID: 38212768 PMCID: PMC10785352 DOI: 10.1186/s12929-024-00999-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Metabolic remodeling and changes in tumor immune microenvironment (TIME) in osteosarcoma are important factors affecting prognosis and treatment. However, the relationship between metabolism and TIME needs to be further explored. METHODS RNA-Seq data and clinical information of 84 patients with osteosarcoma from the TARGET database and an independent cohort from the GEO database were included in this study. The activity of seven metabolic super-pathways and immune infiltration levels were inferred in osteosarcoma patients. Metabolism-related genes (MRGs) were identified and different metabolic clusters and MRG-related gene clusters were identified using unsupervised clustering. Then the TIME differences between the different clusters were compared. In addition, an MRGs-based risk model was constructed and the role of a key risk gene, ST3GAL4, in osteosarcoma cells was explored using molecular biological experiments. RESULTS This study revealed four key metabolic pathways in osteosarcoma, with vitamin and cofactor metabolism being the most relevant to prognosis and to TIME. Two metabolic pathway-related clusters (C1 and C2) were identified, with some differences in immune activating cell infiltration between the two clusters, and C2 was more likely to respond to two chemotherapeutic agents than C1. Three MRG-related gene clusters (GC1-3) were also identified, with significant differences in prognosis among the three clusters. GC2 and GC3 had higher immune cell infiltration than GC1. GC3 is most likely to respond to immune checkpoint blockade and to three commonly used clinical drugs. A metabolism-related risk model was developed and validated. The risk model has strong prognostic predictive power and the low-risk group has a higher level of immune infiltration than the high-risk group. Knockdown of ST3GAL4 significantly inhibited proliferation, migration, invasion and glycolysis of osteosarcoma cells and inhibited the M2 polarization of macrophages. CONCLUSION The metabolism of vitamins and cofactors is an important prognostic regulator of TIME in osteosarcoma, MRG-related gene clusters can well reflect changes in osteosarcoma TIME and predict chemotherapy and immunotherapy response. The metabolism-related risk model may serve as a useful prognostic predictor. ST3GAL4 plays a critical role in the progression, glycolysis, and TIME of osteosarcoma cells.
Collapse
Affiliation(s)
- Changwu Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hong Shen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chao Deng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Christian Kleber
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Georg Osterhoff
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Nikolas Schopow
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
46
|
Rondel F, Farooq H, Hosseini R, Grinshpon M, Zelikovsky A. EMPathways2: Estimation of Enzyme Expression and Metabolic Pathway Activity Using RNA-Seq Reads. Methods Mol Biol 2024; 2812:39-46. [PMID: 39068356 DOI: 10.1007/978-1-0716-3886-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In this chapter, we outline an approach to analyzing metatranscriptomic data, focusing on the assessment of differential enzyme expression and metabolic pathway activities using a novel bioinformatics software tool, EMPathways2. The analysis pipeline commences with raw data originating from a sequencer and concludes with an output of enzyme expressions and an estimate of metabolic pathway activities. The initial step involves aligning specific transcriptomes assembled from RNA-Seq data using Bowtie2 and acquiring gene expression data with IsoEM2. Subsequently, the pipeline proceeds to quality assessment and preprocessing of the input data, ensuring accurate estimates of enzymes and their differential regulation. Upon completion of the preprocessing stage, EMPathways2 is employed to decipher the intricate relationships between genes, enzymes, and pathways. An online repository containing sample data has been made available, alongside custom Python scripts designed to modify the output of the programs within the pipeline for diverse downstream analyses. This chapter highlights the technical aspects and practical applications of using EMPathways2, which facilitates the advancement of transcriptome data analysis and contributes to a deeper understanding of the complex regulatory mechanisms underlying living systems.
Collapse
Affiliation(s)
- Filipp Rondel
- Department of Computer Science, Georgia State University, Atlanta, GA, USA
| | - Hafsa Farooq
- Department of Computer Science, Georgia State University, Atlanta, GA, USA
| | - Roya Hosseini
- Department of Computer Science, Georgia State University, Atlanta, GA, USA
| | - Mark Grinshpon
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| | - Alex Zelikovsky
- Department of Computer Science, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
47
|
Shen L, Zhou P, Wang YM, Zhu Z, Yuan Q, Cao S, Li J. Supramolecular nanoparticles based on elastin-like peptides modified capsid protein as drug delivery platform with enhanced cancer chemotherapy efficacy. Int J Biol Macromol 2024; 256:128107. [PMID: 38007030 DOI: 10.1016/j.ijbiomac.2023.128107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/11/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Cancer, a prevalent disease posing significant threats to human health and longevity, necessitates effective therapeutic interventions. Chemotherapy has emerged as a primary strategy following surgical procedures for combating most malignancies. Despite the considerable efficacy of conventional chemotherapeutic agents against cancer cells, their utility is hindered by profound challenges such as multidrug resistance and deleterious toxic side effects, thereby limiting their systemic application. To tackle these challenges, we have devised a promising nanomedicine platform based on a plant virus. Specifically, we have selected the cowpea melanoma mottled virus (CCMV) as our nano-delivery system owing to its monodisperse and homogeneous size, as well as its intrinsic ability for controlled self-assembly. Leveraging the potential of this platform, we have engineered CCMV-based nanoparticles functionalized with elastin-like peptides (ELPs) at their N-terminal region. The target protein, CP-ELP, was expressed via E.coli, enabling encapsulation of the model drug DOX upon structural domain modification of the protein. The resulting nanoparticles exhibit uniform size distribution, facilitating efficient internalization by tumor cells and subsequent intracellular drug release, leading to enhanced antitumor efficacy. In addition, EVLP@DOX nanoparticles were found to activate immune response of tumor microenvironment in vivo, which further inhibiting tumor growth. Our designed nanoparticles have also demonstrated remarkable therapeutic effectiveness and favorable biological safety profiles in both murine melanoma and colorectal cancer models.
Collapse
Affiliation(s)
- Luxuan Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Min Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhixiong Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuqin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jianshu Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
48
|
Kim HS, Noh YK, Min KW, Kim DH. Low CDKN1B Expression Associated with Reduced CD8+ T Lymphocytes Predicts Poor Outcome in Breast Cancer in a Machine Learning Analysis. J Pers Med 2023; 14:30. [PMID: 38248731 PMCID: PMC10817603 DOI: 10.3390/jpm14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024] Open
Abstract
The cyclin-dependent kinase inhibitor 1B (CDKN1B) gene, which encodes the p27Kip1 protein, is important in regulating the cell cycle process and cell proliferation. Its role in breast cancer prognosis is controversial. We evaluated the significance and predictive role of CDKN1B expression in breast cancer prognosis. We investigated the clinicopathologic factors, survival rates, immune cells, gene sets, and prognostic models according to CDKN1B expression in 3794 breast cancer patients. We performed gene set enrichment analysis (GSEA), in silico cytometry, pathway network analyses, gradient boosting machine (GBM) learning, and in vitro drug screening. High CDKN1B expression levels in breast cancer correlated with high lymphocyte infiltration signature scores and increased CD8+ T cells, both of which were associated with improved prognosis in breast cancer. which were associated with a better prognosis. CDKN1B expression was associated with gene sets for the upregulation of T-cell receptor signaling pathways and downregulation of CD8+ T cells. Pathway network analysis revealed a direct link between CDKN1B and the pathway involved in the positive regulation of the protein catabolic process pathway. In addition, an indirect link was identified between CDKN1B and the T-cell receptor signaling pathway. In in vitro drug screening, BMS-345541 demonstrated efficacy as a therapeutic targeting of CDKN1B, effectively impeding the growth of breast cancer cells characterized by low CDKN1B expression. The inclusion of CDKN1B expression in GBM models increased the accuracy of survival predictions. CDKN1B expression plays a significant role in breast cancer progression, implying that targeting CDKN1B might be a promising strategy for treating breast cancer.
Collapse
Affiliation(s)
- Hyung-Suk Kim
- Division of Breast Surgery, Department of Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 15588, Republic of Korea;
| | - Yung-Kyun Noh
- Department of Computer Science, Hanyang University, 222 Wangsimni-ro, Seoul 04763, Republic of Korea;
- School of Computational Sciences, Korea Institute for Advanced Study, Seoul 02455, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Uijeongbu Eulji Medical Center, School of Medicine, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, School of Medicine, Sungkyunkwan University, 29 Saemunanro, Seoul 03181, Republic of Korea
| |
Collapse
|
49
|
Gao M, Huang J, Yang B, Liu Q, Luo M, Yang B, Li X, Liu X. Identification of efferocytosis-related subtypes in gliomas and elucidating their characteristics and clinical significance. Front Cell Dev Biol 2023; 11:1295891. [PMID: 38161335 PMCID: PMC10757721 DOI: 10.3389/fcell.2023.1295891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: Gliomas, the most prevalent tumors of the central nervous system, are known for their aggressive nature and poor prognosis. The heterogeneity among gliomas leads to varying responses to the same treatments, even among similar glioma types. In our study, we efferocytosis-related subtypes and explored their characteristics in terms of immune landscape, intercellular communication, and metabolic processes, ultimately elucidating their potential clinical implications. Methods and Results: We first identified efferocytosis-related subtypes in Bulk RNA-seq using the NMF algorithm. We then preliminarily demonstrated the correlation of these subtypes with efferocytosis by examining enrichment scores of cell death pathways, macrophage infiltration, and the expression of immune ligands. Our analysis of single-cell RNA-seq data further supported the association of these subtypes with efferocytosis. Through enrichment analysis, we found that efferocytosis-related subtypes differ from other types of gliomas in terms of immune landscape, intercellular communication, and substance metabolism. Moreover, we found that the efferocytosis-related classification is a prognostic factor with robust predictive performance by calculating the AUC values. We also found that efferocytosis-related subtypes, when compared with other gliomas in drug sensitivity, survival, and TIDE scores, show a clear link to the effectiveness of chemotherapy, radiotherapy, and immunotherapy in glioma patients. Discussion: We identified efferocytosis-related subtypes in gliomas by analyzing the expression of 137 efferocytosis-associated genes, exploring their characteristics in immune landscape, intercellular communication, metabolic processes, and genomic variations. Moreover, we discovered that the classification of efferocytosis-related subtypes has a strong prognostic predictive power and holds potential significance in guiding clinical treatment.
Collapse
Affiliation(s)
- Mengge Gao
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Jinsheng Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bo Yang
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Qiong Liu
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Miaoqing Luo
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Biying Yang
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Xujia Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaofang Liu
- Department of Clinical Nutrition, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Chen X, Deng M, Wang Z, Huang C. MMP3C: an in-silico framework to depict cancer metabolic plasticity using gene expression profiles. Brief Bioinform 2023; 25:bbad471. [PMID: 38145946 PMCID: PMC10749788 DOI: 10.1093/bib/bbad471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023] Open
Abstract
Metabolic plasticity enables cancer cells to meet divergent demands for tumorigenesis, metastasis and drug resistance. Landscape analysis of tumor metabolic plasticity spanning different cancer types, in particular, metabolic crosstalk within cell subpopulations, remains scarce. Therefore, we proposed a new in-silico framework, termed as MMP3C (Modeling Metabolic Plasticity by Pathway Pairwise Comparison), to depict tumor metabolic plasticity based on transcriptome data. Next, we performed an extensive metabo-plastic analysis of over 6000 tumors comprising 13 cancer types. The metabolic plasticity within distinct cell subpopulations, particularly interplay with tumor microenvironment, were explored at single-cell resolution. Ultimately, the metabo-plastic events were screened out for multiple clinical applications via machine learning methods. The pilot research indicated that 6 out of 13 cancer types exhibited signs of the Warburg effect, implying its high reliability and robustness. Across 13 cancer types, high metabolic organized heterogeneity was found, and four metabo-plastic subtypes were determined, which link to distinct immune and metabolism patterns impacting prognosis. Moreover, MMP3C analysis of approximately 60 000 single cells of eight breast cancer patients unveiled several metabo-plastic events correlated to tumorigenesis, metastasis and immunosuppression. Notably, the metabolic features screened out by MMP3C are potential biomarkers for diagnosis, tumor classification and prognosis. MMP3C is a practical cross-platform tool to capture tumor metabolic plasticity, and our study unveiled a core set of metabo-plastic pairs among diverse cancer types, which provides bases toward improving response and overcoming resistance in cancer therapy.
Collapse
Affiliation(s)
- Xingyu Chen
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China
| | - Min Deng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Zihan Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China
| | - Chen Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China
| |
Collapse
|