1
|
Luo LZ, Kim JH, Herrera I, Wu S, Wu X, Park SS, Cho J, Cope L, Xian L, West BE, Calderon-Espinosa J, Kim J, Thompson Z, Maloo I, Larman T, Reddy KL, Feng Y, Fearon ER, Sears CL, Resar L. HMGA1 acts as an epigenetic gatekeeper of ASCL2 and Wnt signaling during colon tumorigenesis. J Clin Invest 2025; 135:e184442. [PMID: 39895630 PMCID: PMC11785931 DOI: 10.1172/jci184442] [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: 07/11/2024] [Accepted: 11/27/2024] [Indexed: 02/04/2025] Open
Abstract
Mutated tumor cells undergo changes in chromatin accessibility and gene expression, resulting in aberrant proliferation and differentiation, although how this occurs is unclear. HMGA1 chromatin regulators are abundant in stem cells and oncogenic in diverse tissues; however, their role in colon tumorigenesis is only beginning to emerge. Here, we uncover a previously unknown epigenetic program whereby HMGA1 amplifies Wnt signaling during colon tumorigenesis driven by inflammatory microbiota and/or Adenomatous polyposis coli (Apc) inactivation. Mechanistically, HMGA1 "opens" chromatin to upregulate the stem cell regulator, Ascl2, and downstream Wnt effectors, promoting stem and Paneth-like cell states while depleting differentiated enterocytes. Loss of just one Hmga1 allele within colon epithelium restrains tumorigenesis and Wnt signaling driven by mutant Apc and inflammatory microbiota. However, HMGA1 deficiency has minimal effects in colon epithelium under homeostatic conditions. In human colon cancer cells, HMGA1 directly induces ASCL2 by recruiting activating histone marks. Silencing HMGA1 disrupts oncogenic properties, whereas reexpression of ASCL2 partially rescues these phenotypes. Further, HMGA1 and ASCL2 are coexpressed and upregulated in human colorectal cancer. Together, our results establish HMGA1 as an epigenetic gatekeeper of Wnt signals and cell state under conditions of APC inactivation, illuminating HMGA1 as a potential therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Li Z. Luo
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jung-Hyun Kim
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Iliana Herrera
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shaoguang Wu
- Division of Infectious Diseases, Department of Medicine
| | - Xinqun Wu
- Division of Infectious Diseases, Department of Medicine
| | - Seong-Sik Park
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Juyoung Cho
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Leslie Cope
- Sidney Kimmel Comprehensive Cancer Center, Division of Biostatistics
| | - Lingling Xian
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bailey E. West
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pathobiology Graduate Program, Department of Pathology, and
| | - Julian Calderon-Espinosa
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Human Genetics Graduate Program, Department of Genetics and Molecular Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph Kim
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zanshé Thompson
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Isha Maloo
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biochemistry and Molecular Biology Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Karen L. Reddy
- Department of Biological Chemistry, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ying Feng
- Department of Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric R. Fearon
- Department of Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cynthia L. Sears
- Division of Infectious Diseases, Department of Medicine
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and
- Molecular Immunology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda Resar
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Comprehensive Cancer Center, Division of Biostatistics
- Pathobiology Graduate Program, Department of Pathology, and
- Human Genetics Graduate Program, Department of Genetics and Molecular Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biochemistry and Molecular Biology Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Pathology and
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and
| |
Collapse
|
2
|
Zhao Y, Liu MJ, Zhang L, Yang Q, Sun QH, Guo JR, Lei XY, He KY, Li JQ, Yang JY, Jian YP, Xu ZX. High mobility group A1 (HMGA1) promotes the tumorigenesis of colorectal cancer by increasing lipid synthesis. Nat Commun 2024; 15:9909. [PMID: 39548107 PMCID: PMC11568219 DOI: 10.1038/s41467-024-54400-0] [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: 11/05/2023] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to meet the high energy and biosynthetic demands required for their proliferation. High mobility group A1 (HMGA1) is a structural transcription factor and frequently overexpressed in human colorectal cancer (CRC). Here, we show that HMGA1 promotes CRC progression by driving lipid synthesis in a AOM/DSS-induced CRC mouse model. Using conditional knockout (Hmga1△IEC) and knock-in (Hmga1IEC-OE/+) mouse models, we demonstrate that HMGA1 enhances CRC cell proliferation and accelerates tumor development by upregulating fatty acid synthase (FASN). Mechanistically, HMGA1 increases the transcriptional activity of sterol regulatory element-binding protein 1 (SREBP1) on the FASN promoter, leading to increased lipid accumulation in intestinal epithelial cells. Moreover, a high-fat diet exacerbates CRC progression in Hmga1△IEC mice, while pharmacological inhibition of FASN by orlistat reduces tumor growth in Hmga1IEC-OE/+ mice. Our findings suggest that targeting lipid metabolism could offer a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qi Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
3
|
Li L, Cheng H, Gong L, Huang Y, Yang J, Yan Q, Dai S, Wang J. Cuproptosis/OXPHOS tendency prediction of prognosis and immune microenvironment of esophageal squamous cell carcinoma: Bioinformatics analysis and experimental validation. Gene 2024; 902:148156. [PMID: 38211899 DOI: 10.1016/j.gene.2024.148156] [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: 10/28/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND Cuproptosis is a newly discovered cell death mechanism that relies on mitochondrial respiration, for which oxidative phosphorylation (OXPHOS) is an essential part. However, the detailed mechanisms of cuproptosis associated with OXPHOS in esophageal squamous cell carcinoma (ESCC) and how this correlation affects prognosis still remains unclear. METHODS scRNA-seq data of ESCC were downloaded from SRA (Sequence Read Archive) database. "AUCell" algorithm was used to grouping epithelial cells according to cuproptosis and OXPHOS score. Cell-cell communication, Pseudo-time Trajectory and transcription factor enrichment analysis were repectively conducted by "CellChat", "monocle3" package and "pySCENIC" algorithm. Univariate and LASSO cox regression analysis were used to construct the prognostic cuproptosis-OXPHOS signature. Finally, CCK-8 assay and DCFH-DA staining assay were respectively validated the sensitive and ROS production of elesclomol. RESULTS scRNA-seq data were analyzed to identify 10 core cell types. According to the median scores for cuproptosis and OXPHOS, malignant epithelial cells were divided into double high, double low, and mixed groups. The double high group distributed at the end of the pseudo-time trajectory and harbored HMGA1(+) as specific transcriptional regulons. Knockdown of HMGA1 partly reversed the inhibition of cell viability visualized by CCK-8 assay, while reactive oxygen species (ROS) production by elesclomol was enhanced after HMGA1 silencing. Furthermore, the immunosuppressive signal was significantly increased in the double high group detected by 'CellChat' in single-cell data and 'ssGSEA' in bulk data followed by 'CIBERSORTx' algorithm. Finally, a new cuproptosis-OXPHOS prognostic signature (CNN2, ATP6V0E1, PSMD6, CCDC25, IGFBP2, MT1E, and RPS4Y1) was constructed for the prediction of the prognosis, and a high-risk group corresponding to a more sensitive tendency to erlotinib, dasatinib, and bosutinib treatment was identified. CONCLUSIONS Our study revealed the relationship between OXPHOS and tendency of cuproptosis in ESCC, and malignant cells with this characteristic exerted immunosuppressive signals and indicated poor prognosis. Furthermore, we constructed the regulatory network in high cuproptosis-OXPHOS ESCC and identified HMGA1 as a potential regulator molecule of cuproptosis mediated by elesclomol.
Collapse
Affiliation(s)
- Liang Li
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China
| | - Haiyan Cheng
- Department of Gynecology, The Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong 266042, PR China
| | - Li Gong
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China
| | - Yongcheng Huang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China
| | - Jie Yang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China
| | - Qihang Yan
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China
| | - Shuqin Dai
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China.
| | - Junye Wang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510030, PR China.
| |
Collapse
|
4
|
Wang Q, Chen F, He Y, Gao Y, Wang J, Chu S, Xie P, Zhong J, Shan H, Bai J, Hou P. Polypyrimidine tract-binding protein 3/insulin-like growth factor 2 mRNA-binding proteins 3/high-mobility group A1 axis promotes renal cancer growth and metastasis. iScience 2024; 27:109158. [PMID: 38405614 PMCID: PMC10884747 DOI: 10.1016/j.isci.2024.109158] [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: 09/11/2023] [Revised: 01/05/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024] Open
Abstract
Polypyrimidine tract-binding protein 3 (PTBP3) plays an important role in the post-transcriptional regulation of gene expression, including mRNA splicing, translation, and stability. Increasing evidence has shown that PTBP3 promotes cancer progression in several tumor types. However, the molecular mechanisms of PTBP3 in renal cell carcinoma (RCC) remain unknown. Here, tissue microarrays (TMAs) suggested that PTBP3 expression was increased in human RCC and that high PTBP3 expression was correlated with poor five-year overall survival and disease-free survival. We also showed that PTBP3 binds with HMGA1 mRNA in the 3'UTR region and let-7 miRNAs. PTBP3 interacted with IGF2BP3, and the PTBP3/IGF2BP3 axis prevented let-7 mediated HMGA1 mRNA silencing. PTBP3 promotes renal cancer cell growth and metastasis in vitro and in vivo. Taken together, our findings indicate PTBP3 serves as a regulator of HMGA1 and suggest its potential as a therapeutic agent for RCC.
Collapse
Affiliation(s)
- Qianqing Wang
- Department of Gynecology Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Fang Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yu He
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yue Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jiawen Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Pei Xie
- Department of Gynecology Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Jiateng Zhong
- Department of Gynecology Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Haixia Shan
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Jin Bai
- Department of Gynecology Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan 453000, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Pingfu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
5
|
Wang J, Zhang X, Wang X, Li F, Zhang D, Li X, Zhang Y, Zhao Y, Song Q, Zhao L, Xu D, Cheng J, Li W, Zhou B, Lin C, Wang W. Polymorphism and expression of the HMGA1 gene and association with tail fat deposition in Hu sheep. Anim Biotechnol 2023; 34:1626-1634. [PMID: 34775926 DOI: 10.1080/10495398.2021.1998093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hu sheep is an excellent short fat-tailed breed in China. Fat deposition in Hu sheep tail affects carcass quality and consumes a lot of energy, leading to an increase in feed cost. The objective of this study was to analyze the effects of HMGA1 polymorphism on tail fat weight in Hu sheep. Partial coding and non-coding sequences of HMGA1 were amplified with PCR and single nucleotide polymorphisms (SNP) of HMGA1 in 1163 Hu sheep were detected using DNA sequencing and KASPar technology. RT-qPCR analysis was performed to test HMGA1 expression in different tissues. The results showed that the expression of HMGA1 was higher in the duodenum, liver, spleen, kidney, and lung than in the heart, muscle, rumen, tail fat, and lymph. A mutation, g.5312 C > T, was detected in HMGA1; g.5312 C > T was significantly associated with tail fat weight, relative weight of tail fat (body weight), and relative weight of tail fat (carcass) (p < 0.05). The tail fat weight of the TT genotype was remarkably higher than that of the CC and TC genotypes. Therefore, HMGA1 can be used as a genetic marker for marker-assisted selection of tail fat weight in Hu sheep.
Collapse
Affiliation(s)
- Jianghui Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiaoxue Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiaojuan Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Fadi Li
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
- Engineering Laboratory of Sheep Breeding and Reproduction Biotechnology in Gansu Province, Minqin, China
| | - Deyin Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiaolong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yukun Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yuan Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Qizhi Song
- Linze County Animal Disease Prevention and Control Center of Gansu Province, Linze, China
| | - Liming Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Dan Xu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jiangbo Cheng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Wenxin Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Bubo Zhou
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Changchun Lin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Weimin Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Jia J, Guo P, Zhang L, Kong W, Wang F. LINC01614 Promotes Colorectal Cancer Cell Growth and Migration by Regulating miR-217-5p/HMGA1 Axis. Anal Cell Pathol (Amst) 2023; 2023:6833987. [PMID: 39282156 PMCID: PMC11401691 DOI: 10.1155/2023/6833987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 09/18/2024] Open
Abstract
Colorectal cancer (CRC) substantially contributes to cancer-related deaths worldwide. Recently, a long non-coding RNA (lncRNA), LINC01614, has emerged as a vital gene regulator in cancer progression. Yet, how LINC01614 affects CRC progression remains enigmatic. Here, we defined LINC01614 expression in CRC, investigated the performance of CRC cells lacking LINC01614, and elucidated the underpinning mechanism. We observed that LINC01614 was upregulated in both CRC tissues and cell lines. LINC01614 knockdown repressed the proliferation and metastasis capacity of CRC cell lines. Consistently, an in vivo LINC01614 deficiency model exhibited slow tumor growth rate. Moreover, luciferase reporter assay, RNA pull-down, and immunoprecipitation confirmed that LINC01614 targeted miR-217-5p. LINC01614 knockdown reduced the expression of HMGA1 and N-cadherin, while increasing that of E-cadherin, resulting in decreased viability, proliferation, migration, and invasion capacity of CRC cells. Our results demonstrate that LINC01614 regulates CRC progression by modulating the miR-217-5p/HMGA1 axis, thus holding great potential as a prognostic biomarker for CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Jiwei Jia
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Pei Guo
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Li Zhang
- Department of Pathology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Wenqing Kong
- Central Ward Operating Room, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Fangfang Wang
- Outpatient Operating Room, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| |
Collapse
|
7
|
Mason SE, Manoli E, Alexander JL, Poynter L, Ford L, Paizs P, Adebesin A, McKenzie JS, Rosini F, Goldin R, Darzi A, Takats Z, Kinross JM. Lipidomic Profiling of Colorectal Lesions for Real-Time Tissue Recognition and Risk-Stratification Using Rapid Evaporative Ionization Mass Spectrometry. Ann Surg 2023; 277:e569-e577. [PMID: 34387206 DOI: 10.1097/sla.0000000000005164] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Rapid evaporative ionization mass spectrometry (REIMS) is a metabolomic technique analyzing tissue metabolites, which can be applied intraoperatively in real-time. The objective of this study was to profile the lipid composition of colorectal tissues using REIMS, assessing its accuracy for real-time tissue recognition and risk-stratification. SUMMARY BACKGROUND DATA Metabolic dysregulation is a hallmark feature of carcinogenesis; however, it remains unknown if this can be leveraged for real-time clinical applications in colorectal disease. METHODS Patients undergoing colorectal resection were included, with carcinoma, adenoma and paired-normal mucosa sampled. Ex vivo analysis with REIMS was conducted using monopolar diathermy, with the aerosol aspirated into a Xevo G2S QToF mass spectrometer. Negatively charged ions over 600 to 1000 m/z were used for univariate and multivariate functions including linear discriminant analysis. RESULTS A total of 161 patients were included, generating 1013 spectra. Unique lipidomic profiles exist for each tissue type, with REIMS differentiating samples of carcinoma, adenoma, and normal mucosa with 93.1% accuracy and 96.1% negative predictive value for carcinoma. Neoplasia (carcinoma or adenoma) could be predicted with 96.0% accuracy and 91.8% negative predictive value. Adenomas can be risk-stratified by grade of dysplasia with 93.5% accuracy, but not histological subtype. The structure of 61 lipid metabolites was identified, revealing that during colorectal carcinogenesis there is progressive increase in relative abundance of phosphatidylglycerols, sphingomyelins, and mono-unsaturated fatty acid-containing phospholipids. CONCLUSIONS The colorectal lipidome can be sampled by REIMS and leveraged for accurate real-time tissue recognition, in addition to riskstratification of colorectal adenomas. Unique lipidomic features associated with carcinogenesis are described.
Collapse
Affiliation(s)
- Sam E Mason
- Department of Surgery and Cancer, Imperial College, London
| | | | - James L Alexander
- Department of Metabolism, Digestion and Reproduction, Imperial College, London; and
| | - Liam Poynter
- Department of Surgery and Cancer, Imperial College, London
| | - Lauren Ford
- Department of Surgery and Cancer, Imperial College, London
| | - Petra Paizs
- Department of Metabolism, Digestion and Reproduction, Imperial College, London; and
| | - Afeez Adebesin
- Department of Surgery and Cancer, Imperial College, London
| | - James S McKenzie
- Department of Metabolism, Digestion and Reproduction, Imperial College, London; and
| | | | - Rob Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College, London; and
| | - Ara Darzi
- Department of Surgery and Cancer, Imperial College, London
| | - Zoltan Takats
- Department of Metabolism, Digestion and Reproduction, Imperial College, London; and
| | | |
Collapse
|
8
|
Zhu Y, Yang Y, Bu H, Huang H, Chen H, Ran J, Qin L, Ni Y, Yao M, Song T, Li M, Yang Y, Guo T, Chao N, Liu Z, Li W, Zhang L. Apelin‐mediated deamidation of
HMGA1
promotes tumorigenesis by enhancing
SREBP1
activity and lipid synthesis. Cancer Sci 2022; 113:3722-3734. [PMID: 36087034 PMCID: PMC9633285 DOI: 10.1111/cas.15515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 02/05/2023] Open
Abstract
Enhanced fatty acid synthesis provides proliferation and survival advantages for tumor cells. Apelin is an adipokine, which serves as a ligand of G protein–coupled receptors that promote tumor growth in malignant cancers. Here, we confirmed that apelin increased sterol regulatory element–binding protein 1 (SREBP1) activity and induced the expression of glutamine amidotransferase for deamidating high‐mobility group A 1 (HMGA1) to promote fatty acid synthesis and proliferation of lung cancer cells. This post‐translational modification stabilized the HMGA1 expression and enhanced the formation of the apelin‐HMGA1‐SREBP1 complex to facilitate SREBP1 activity for lipid metabolism and lung cancer cell growth. We uncovered the pivotal role of apelin‐mediated deamidation of HMGA1 in lipid metabolism and tumorigenesis of lung cancer cells.
Collapse
Affiliation(s)
- Yihan Zhu
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital Sichuan University Chengdu China
| | - Ying Yang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Hong Bu
- Department of Pathology, West China Hospital Sichuan University Chengdu China
| | - Hong Huang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital Sichuan University Chengdu China
| | - Hongyu Chen
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital Sichuan University Chengdu China
| | - Jingjing Ran
- Laboratory of Human Diseases and Immunotherapies, West China Hospital Sichuan University Chengdu China
| | - Liwen Qin
- Administration of Research Park, West China Hospital Sichuan University Chengdu China
| | - Yinyun Ni
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Menglin Yao
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Tingting Song
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Mufeng Li
- Department of Nuclear Medicine, West China Hospital Sichuan University Chengdu China
| | - Yongfeng Yang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Tingting Guo
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Ningning Chao
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Zhiqing Liu
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu China
| |
Collapse
|
9
|
Güçlü E, Eroğlu Güneş C, Kurar E, Vural H. Knockdown of lncRNA HIF1A-AS2 increases drug sensitivity of SCLC cells in association with autophagy. Med Oncol 2021; 38:113. [PMID: 34378101 DOI: 10.1007/s12032-021-01562-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
The aim of this study was to determine the effect of lncRNA HIF1A-AS2 on autophagy-associated drug resistance in small cell lung cancer (SCLC) cells. The expression of HIF1A-AS2 was silenced by siRNA in doxorubicin-sensitive H69 and doxorubicin-resistant H69AR cells. Then, cytotoxicity, apoptosis and autophagy analyses were carried out in the normoxic and CoCl2-induced hypoxic environment. The effect of HIF1A-AS2 on the expression levels of genes, which are associated with drug resistance and autophagy, was determinated by qRT-PCR analysis. The levels of MRP1, HIF-1α and Beclin-1 were analyzed by western blot method. Knockdown of HIF1A-AS2 increased doxorubicin sensitivity of SCLC cells and decreased autophagy. Knockdown of HIF1A-AS2 has also affected the expression of several genes that will increase drug sensitivity and inhibit autophagy in both cell lines. The levels of HIF-1α and Beclin-1 were decreased in both cell lines by knockdown of HIF1A-AS2. MRP1 expression was decrease in H69AR cells. In addition, CoCl2-induced hypoxic environment decreased in doxorubicin sensitivity of H69 cells, and knockdown of HIF1A-AS2 reversed this effect of hypoxia. Knockdown of HIF1A-AS2 increased drug sensitivity of SCLC cells in relation to autophagy. Therefore, hypoxia-HIF1A-AS2-autophagy interaction is thought to be determinative in drug sensitivity of these cells.
Collapse
Affiliation(s)
- Ebru Güçlü
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey.
| | - Canan Eroğlu Güneş
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ercan Kurar
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hasibe Vural
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
10
|
Matsubara K, Matsubara Y, Uchikura Y, Takagi K, Yano A, Sugiyama T. HMGA1 Is a Potential Driver of Preeclampsia Pathogenesis by Interference with Extravillous Trophoblasts Invasion. Biomolecules 2021; 11:biom11060822. [PMID: 34072941 PMCID: PMC8227282 DOI: 10.3390/biom11060822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia (PE) is a serious disease that can be fatal for the mother and fetus. The two-stage theory has been proposed as its cause, with the first stage comprising poor placentation associated with the failure of fertilized egg implantation. Successful implantation and placentation require maternal immunotolerance of the fertilized egg as a semi-allograft and appropriate extravillous trophoblast (EVT) invasion of the decidua and myometrium. The disturbance of EVT invasion during implantation in PE results in impaired spiral artery remodeling. PE is thought to be caused by hypoxia during remodeling failure-derived poor placentation, which results in chronic inflammation. High-mobility group protein A (HMGA) is involved in the growth and invasion of cancer cells and likely in the growth and invasion of trophoblasts. Its mechanism of action is associated with immunotolerance. Thus, HMGA is thought to play a pivotal role in successful pregnancy, and its dysfunction may be related to the pathogenesis of PE. The evaluation of HMGA function and its changes in PE might confirm that it is a reliable biomarker of PE and provide prospects for PE treatment through the induction of EVT proliferation and invasion during the implantation.
Collapse
Affiliation(s)
- Keiichi Matsubara
- Department of Regional Pediatrics and Perinatology, Graduate School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan
- Correspondence:
| | - Yuko Matsubara
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Yuka Uchikura
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Katsuko Takagi
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Akiko Yano
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| |
Collapse
|
11
|
Liu H, Zhang X, Jin X, Yang Y, Liang G, Ma Y, Wang B. Long Noncoding RNA VPS9D1-AS1 Sequesters microRNA-525-5p to Promote the Oncogenicity of Colorectal Cancer Cells by Upregulating HMGA1. Cancer Manag Res 2020; 12:9915-9928. [PMID: 33116849 PMCID: PMC7553769 DOI: 10.2147/cmar.s273687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/03/2020] [Indexed: 12/24/2022] Open
Abstract
Background The long noncoding RNA VPS9D1 antisense RNA 1 (VPS9D1-AS1) has emerged as a critical regulator in non-small-cell lung, gastric, and prostate cancers. In this study, we measured the expression levels of VPS9D1-AS1 in colorectal cancer (CRC) and determined the role of VPS9D1-AS1 in regulating the biological activities of CRC cells. In addition, we thoroughly elucidated the molecular mechanism mediating the oncogenic activities of VPS9D1-AS1 in CRC. Methods The expression levels of VPS9D1-AS1 in CRC tissues and cell lines were detected via quantitative reverse transcription-polymerase chain reaction. Loss-of-function experiments were performed to detect the effects of VPS9D1-AS1 silencing on CRC cell proliferation, apoptosis, migration, and invasion as well as on tumor growth in vivo. Bioinformatics analysis predicted the potential microRNAs (miRNAs) interacting with VPS9D1-AS1, and this prediction was further confirmed via RNA immunoprecipitation and luciferase reporter assays. Results Our results demonstrated the upregulated expression of VPS9D1-AS1 in CRC tissues and cell lines. Functionally, VPS9D1-AS1 interference suppressed CRC cell proliferation, migration, and invasion and promoted cell apoptosis in vitro. In addition, the loss of VPS9D1-AS1 hindered tumor growth in vivo. Mechanistic studies identified VPS9D1-AS1 as a competing endogenous RNA in CRC cells, in which VPS9D1-AS1 acted as a molecular sponge of miR-525-5p and consequently increased the expression of high-mobility group AT-hook 1 (HMGA1). Moreover, rescue experiments revealed that the regulatory effects of VPS9D1-AS1 deficiency on CRC cells were abolished after miR-525-5p inhibition or HMGA1 restoration. Conclusion The newly identified competing endogenous RNA pathway involving VPS9D1-AS1, miR-525-5p, and HMGA1 is implicated in the control of CRC progression and may provide an effective target for CRC diagnosis and therapy.
Collapse
Affiliation(s)
- Hairui Liu
- Department of Abdominal Surgery, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| | - Xueying Zhang
- Department of Oncology Medicine, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| | - Xianmei Jin
- Department of Childhood Solid Tumor, First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Yubo Yang
- Department of Abdominal Surgery, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| | - Guodong Liang
- Department of Abdominal Surgery, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| | - Yuehan Ma
- Department of Abdominal Surgery, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| | - Bing Wang
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, Jilin 130021, People's Republic of China
| |
Collapse
|
12
|
The Role of Non-Coding RNAs in Uveal Melanoma. Cancers (Basel) 2020; 12:cancers12102944. [PMID: 33053887 PMCID: PMC7600503 DOI: 10.3390/cancers12102944] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The development of uveal melanoma is a multifactorial and multi-step process, in which abnormal gene expression plays a key role. Recently, several studies have highlighted the role of non-coding RNAs in the progression of uveal melanoma by affecting different signaling pathways. As important agents in the regulation of genes, non-coding RNAs have enormous potential to open up therapeutic pathways, predict response to treatment, and anticipate patient outcome for uveal melanoma. This review aims to provide a comprehensive view of what we know about ncRNAs in uveal melanoma currently. Abstract Uveal melanoma (UM) is the most common primary intraocular tumor in adulthood. Approximately 50% of patients develop metastatic disease, which typically affects the liver and is usually fatal within one year. This type of cancer is heterogeneous in nature and is divided into two broad groups of tumors according to their susceptibility to develop metastasis. In the last decade, chromosomal abnormalities and the aberrant expression of several signaling pathways and oncogenes in uveal melanomas have been described. Recently, importance has been given to the association of the mentioned deregulation with the expression of non-coding RNAs (ncRNAs). Here, we review the different classes of ncRNAs—such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs)—and their contribution to the development of UM. Special attention is given to miRNAs and their regulatory role in physiopathology and their potential as biomarkers. As important agents in gene regulation, ncRNAs have a huge potential for opening up therapeutic pathways, predicting response to treatment, and anticipating patient outcome for UM.
Collapse
|
13
|
Yang M, Guo Y, Liu X, Liu N. HMGA1 Promotes Hepatic Metastasis of Colorectal Cancer by Inducing Expression of Glucose Transporter 3 (GLUT3). Med Sci Monit 2020; 26:e924975. [PMID: 32989212 PMCID: PMC7532698 DOI: 10.12659/msm.924975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common cancers worldwide, and more than half of CRC patients have CRC liver metastasis (CRCLM). Mounting evidence indicates that high mobility group protein A1(HMGA1) is overexpressed in many cancer types, but its role in CRCLM has been obscure. Material/Methods Using immunohistochemistry, we assessed the expression of HMGA1 in 73 patients with CRCLM, and compared HMGA1 mRNA in 17 pairs of CRCs, CRCLM tissues, and normal liver tissues. The clinical significance of HMGA1 was evaluated by analyzing its correlation with the clinicopathological factors and overall survival (OS) rates. The function of HMGA1 in CRC invasion was investigated and the underlying mechanism of HMGA1-induced invasion was explored with in vitro experiments. Results In CRCLMs, the high-HMGA1 and low-HMGA1 patients accounted for 53.42% and 46.58% of all patients, respectively. High HMGA1 expression in CRCLM was significantly associated with low OS rates. In vitro experiments demonstrated that HMGA1 promoted glucose transporter 3 (GLUT3) transcription and expression in CRC cells. GLUT3 was required in HMGA1-involved invasion, and GLUT3 expression was associated with poor prognosis of CRCLM. Conclusions High HMGA1 and GLUT3 expression in CRCLM was significantly correlated with poor prognosis of CRCLM. HMGA1 promoted CRC invasion by elevating GLUT3 transcription and expression.
Collapse
Affiliation(s)
- Meijing Yang
- Department of Geriatrics, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Yang Guo
- Department of Geriatrics, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Xiaoyun Liu
- Department of Cardiology, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Naiqing Liu
- Department of General Surgery, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| |
Collapse
|
14
|
High Mobility Group A (HMGA): Chromatin Nodes Controlled by a Knotty miRNA Network. Int J Mol Sci 2020; 21:ijms21030717. [PMID: 31979076 PMCID: PMC7038092 DOI: 10.3390/ijms21030717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.
Collapse
|
15
|
The Prominent Role of HMGA Proteins in the Early Management of Gastrointestinal Cancers. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2059516. [PMID: 31737655 PMCID: PMC6815579 DOI: 10.1155/2019/2059516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022]
Abstract
GI tumors represent a heterogeneous group of neoplasms concerning their natural history and molecular alterations harbored. Nevertheless, these tumors share very high incidence and mortality rates worldwide and patients' poor prognosis. Therefore, the identification of specific biomarkers could increase the development of personalized medicine, in order to improve GI cancer management. In this sense, HMGA family members (HMGA1 and HMGA2) comprise an important group of genes involved in the genesis and progression of malignant tumors. Additionally, it has also been reported that HMGA1 and HMGA2 display an important role in the detection and progression of GI tumors. In this way, HMGA family members could be used as reliable biomarkers able to efficiently track not only the tumor per se but also the main risk conditions related with their development of GI cancers in the future. Finally, it shall be a promising option to revert the current scenario, once HMGA genes and proteins could represent a convergence point in the complex landscape of GI tumors.
Collapse
|
16
|
HMGA1 exacerbates tumor progression by activating miR-222 through PI3K/Akt/MMP-9 signaling pathway in uveal melanoma. Cell Signal 2019; 63:109386. [PMID: 31394192 DOI: 10.1016/j.cellsig.2019.109386] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/03/2019] [Accepted: 08/04/2019] [Indexed: 12/26/2022]
Abstract
High-mobility group A1 (HMGA1), an architectural transcription factor, participates in different human tumors' biological progression. HMGA1 overexpression is associated with malignant cellular behavior in a wide range of cancers but the underlying mechanism remains poorly illuminated. In this study, we showed PI3K/Akt/MMP9 pathway activity could be positively regulated by HMGA1 using western blotting, real-time polymerase chain reaction (RT-PCR) and immunochemistry both in vitro (C918 and MUM-2B cell lines) and in vivo (xenograft mouse model). Later, MiRTarBase was used to identify the relationship between HMGA1 and miR-222-3p, we found miR-222 is positively regulated by HMGA1. Moreover, the proliferation and migration of UM cells significantly increased in the miR-222 mimics group and decreased in the miR-222 inhibitor group detected by the Annexin V-FITC apoptosis detection kit, CCK-8 and scratch wound-healing. The p-PI3K, p-Akt and MMP9 expressions were elevated in UM cells transfected with miR-222 mimics, and suppressed in the miR-222 inhibitor group. Together, our study highlights that HMGA1 acts as a pivotal regulator in UM tumor growth, proposing a critical viewpoint that HMGA1 expedites progression through the PI3K/Akt/MMP9 pathway and oncogenic miR-222 in UM.
Collapse
|
17
|
Wang Y, Hu L, Zheng Y, Guo L. HMGA1 in cancer: Cancer classification by location. J Cell Mol Med 2019; 23:2293-2302. [PMID: 30614613 PMCID: PMC6433663 DOI: 10.1111/jcmm.14082] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 07/19/2018] [Accepted: 11/16/2018] [Indexed: 12/23/2022] Open
Abstract
The high mobility group A1 (HMGA1) gene plays an important role in numerous malignant cancers. HMGA1 is an oncofoetal gene, and we have a certain understanding of the biological function of HMGA1 based on its activities in various neoplasms. As an architectural transcription factor, HMGA1 remodels the chromatin structure and promotes the interaction between transcriptional regulatory proteins and DNA in different cancers. Through analysis of the molecular mechanism of HMGA1 and clinical studies, emerging evidence indicates that HMGA1 promotes the occurrence and metastasis of cancer. Within a similar location or the same genetic background, the function and role of HMGA1 may have certain similarities. In this paper, to characterize HMGA1 comprehensively, research on various types of tumours is discussed to further understanding of the function and mechanism of HMGA1. The findings provide a more reliable basis for classifying HMGA1 function according to the tumour location. In this review, we summarize recent studies related to HMGA1, including its structure and oncogenic properties, its major functions in each cancer, its upstream and downstream regulation associated with the tumourigenesis and metastasis of cancer, and its potential as a biomarker for clinical diagnosis of cancer.
Collapse
Affiliation(s)
- Yuhong Wang
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| | - Lin Hu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yushuang Zheng
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| | - Lingchuan Guo
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| |
Collapse
|
18
|
Qi C, Cao J, Li M, Liang C, He Y, Li Y, Li J, Zheng X, Wang L, Wei B. HMGA1 Overexpression is Associated With the Malignant Status and Progression of Breast Cancer. Anat Rec (Hoboken) 2018; 301:1061-1067. [PMID: 29316384 DOI: 10.1002/ar.23777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/20/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022]
Abstract
Breast cancer is the most common malignant tumor among women, and the incidence and mortality of breast cancer has rapidly increased in recent years. Studies have indicated that high mobility group A1 (HMGA1), an important member of the HMGA family, plays a role in the pathogenesis and progression of malignant tumors, including breast cancer. This study aims to evaluate the effect of HMGA1 in breast cancer. Interestingly, we found that HMGA1 expression was significantly higher in breast cancer tissues than in adenoma tissues and closely correlated with the clinical stage and histological grade in breast cancer patients. Further study showed that HMGA1 knockdown inhibited the proliferation and migration of breast cancer cells. Thus, the results demonstrated that HMGA1 could act as an independent prognostic indicator in breast cancer. HMGA1 expression was closely correlated with the clinical stage, histological grade, and tumor size in breast cancer patients and breast cancer progression in transgenic MMTV-PyMT mice. Anat Rec, 301:1061-1067, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cuiling Qi
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jinghua Cao
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mengshi Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chenghua Liang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Yajun He
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuanyuan Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jialin Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoming Zheng
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Lijing Wang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bo Wei
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
19
|
Zhang S, Lei R, Wu J, Shan J, Hu Z, Chen L, Ren X, Yao L, Wang J, Wang X. Role of high mobility group A1 and body mass index in the prognosis of patients with breast cancer. Oncol Lett 2017; 14:5719-5726. [PMID: 29113200 PMCID: PMC5661362 DOI: 10.3892/ol.2017.6963] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
The high mobility group A1 (HMGA1) protein is associated with poor prognosis in patients with a wide range of cancers. However, the affect of HMGA1 on the risk of mortality from breast cancer (BC) has not been fully characterized. In the present retrospective multiple center study, the HMGA1 expression level was determined by performing immunohistochemistry on surgical tissue samples of 273 BC specimens from the Second Affiliated Hospital of Zhejiang University (Zhejiang, China) and 310 BCs from the National Engineering Center for Biochip (Shanghai, China). Kaplan-Meier analysis and Cox proportional hazard model were employed to analyze the survivability. HMGA1 expression was significantly associated with tumor histological degree and body mass index (BMI). However, HMGA1 expression showed no prognostic value in patients with BC. Combined evaluation of HMGA1 expression and high BMI (≥24 kg/m2) predicted worse overall survival of BC. Therefore, HMGA1 and BMI were considered to serve synergistic roles in the development and progression of BC, and combined evaluation of HMGA1 expression and high BMI may be an effective marker in predicting poor prognosis of BC patients.
Collapse
Affiliation(s)
- Shizhen Zhang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Rui Lei
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jingjing Wu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jinlan Shan
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zujian Hu
- Department of Breast Surgery, Hangzhou Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lirong Chen
- Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xingchang Ren
- Department of Pathology, Hangzhou Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lifang Yao
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jian Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaochen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
20
|
Sumter TF, Xian L, Huso T, Koo M, Chang YT, Almasri TN, Chia L, Inglis C, Reid D, Resar LMS. The High Mobility Group A1 (HMGA1) Transcriptome in Cancer and Development. Curr Mol Med 2016; 16:353-93. [PMID: 26980699 DOI: 10.2174/1566524016666160316152147] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 02/15/2016] [Accepted: 03/10/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND & OBJECTIVES Chromatin structure is the single most important feature that distinguishes a cancer cell from a normal cell histologically. Chromatin remodeling proteins regulate chromatin structure and high mobility group A (HMGA1) proteins are among the most abundant, nonhistone chromatin remodeling proteins found in cancer cells. These proteins include HMGA1a/HMGA1b isoforms, which result from alternatively spliced mRNA. The HMGA1 gene is overexpressed in cancer and high levels portend a poor prognosis in diverse tumors. HMGA1 is also highly expressed during embryogenesis and postnatally in adult stem cells. Overexpression of HMGA1 drives neoplastic transformation in cultured cells, while inhibiting HMGA1 blocks oncogenic and cancer stem cell properties. Hmga1 transgenic mice succumb to aggressive tumors, demonstrating that dysregulated expression of HMGA1 causes cancer in vivo. HMGA1 is also required for reprogramming somatic cells into induced pluripotent stem cells. HMGA1 proteins function as ancillary transcription factors that bend chromatin and recruit other transcription factors to DNA. They induce oncogenic transformation by activating or repressing specific genes involved in this process and an HMGA1 "transcriptome" is emerging. Although prior studies reveal potent oncogenic properties of HMGA1, we are only beginning to understand the molecular mechanisms through which HMGA1 functions. In this review, we summarize the list of putative downstream transcriptional targets regulated by HMGA1. We also briefly discuss studies linking HMGA1 to Alzheimer's disease and type-2 diabetes. CONCLUSION Further elucidation of HMGA1 function should lead to novel therapeutic strategies for cancer and possibly for other diseases associated with aberrant HMGA1 expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - L M S Resar
- Department of Medicine, Faculty of the Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, MD 21205-2109, USA.
| |
Collapse
|
21
|
Comment on 'MicroRNA-214 suppresses growth, migration and invasion through a novel target, high mobility group AT-hook 1, in human cervical and colorectal cancer cells'. Br J Cancer 2016; 116:e7. [PMID: 27997527 PMCID: PMC5294474 DOI: 10.1038/bjc.2016.409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
22
|
Williams MD, Xian L, Huso T, Park JJ, Huso D, Cope LM, Gang DR, Siems WF, Resar L, Reeves R, Hill HH. Fecal Metabolome in Hmga1 Transgenic Mice with Polyposis: Evidence for Potential Screen for Early Detection of Precursor Lesions in Colorectal Cancer. J Proteome Res 2016; 15:4176-4187. [PMID: 27696867 DOI: 10.1021/acs.jproteome.6b00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Because colorectal cancer (CRC) remains a leading cause of cancer mortality worldwide, more accessible screening tests are urgently needed to identify early stage lesions. We hypothesized that highly sensitive, metabolic profile analysis of stool samples will identify metabolites associated with early stage lesions and could serve as a noninvasive screening test. We therefore applied traveling wave ion mobility mass spectrometry (TWIMMS) coupled with ultraperformance liquid chromatography (UPLC) to investigate metabolic aberrations in stool samples in a transgenic model of premalignant polyposis aberrantly expressing the gene encoding the high mobility group A (Hmga1) chromatin remodeling protein. Here, we report for the first time that the fecal metabolome of Hmga1 mice is distinct from that of control mice and includes metabolites previously identified in human CRC. Significant alterations were observed in fatty acid metabolites and metabolites associated with bile acids (hypoxanthine xanthine, taurine) in Hmga1 mice compared to controls. Surprisingly, a marked increase in the levels of distinctive short, arginine-enriched, tetra-peptide fragments was observed in the transgenic mice. Together these findings suggest that specific metabolites are associated with Hmga1-induced polyposis and abnormal proliferation in intestinal epithelium. Although further studies are needed, these data provide a compelling rationale to develop fecal metabolomic analysis as a noninvasive screening tool to detect early precursor lesions to CRC in humans.
Collapse
Affiliation(s)
- Michael D Williams
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Lingling Xian
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Tait Huso
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Jeong-Jin Park
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - David Huso
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Leslie M Cope
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - David R Gang
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - William F Siems
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Linda Resar
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Raymond Reeves
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Herbert H Hill
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| |
Collapse
|
23
|
Hillion J, Roy S, Heydarian M, Cope L, Xian L, Koo M, Luo LZ, Kellyn K, Ronnett BM, Huso T, Armstrong D, Reddy K, Huso DL, Resar LMS. The High Mobility Group A1 (HMGA1) gene is highly overexpressed in human uterine serous carcinomas and carcinosarcomas and drives Matrix Metalloproteinase-2 (MMP-2) in a subset of tumors. Gynecol Oncol 2016; 141:580-587. [PMID: 27001612 DOI: 10.1016/j.ygyno.2016.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 03/06/2016] [Accepted: 03/16/2016] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Although uterine cancer is the fourth most common cause for cancer death in women worldwide, the molecular underpinnings of tumor progression remain poorly understood. The High Mobility Group A1 (HMGA1) gene is overexpressed in aggressive cancers and high levels portend adverse outcomes in diverse tumors. We previously reported that Hmga1a transgenic mice develop uterine tumors with complete penetrance. Because HMGA1 drives tumor progression by inducing MatrixMetalloproteinase (MMP) and other genes involved in invasion, we explored the HMGA1-MMP-2 pathway in uterine cancer. METHODS To investigate MMP-2 in uterine tumors driven by HMGA1, we used a genetic approach with mouse models. Next, we assessed HMGA1 and MMP-2 expression in primary human uterine tumors, including low-grade carcinomas (endometrial endometrioid) and more aggressive tumors (endometrial serous carcinomas, uterine carcinosarcomas/malignant mesodermal mixed tumors). RESULTS Here, we report for the first time that uterine tumor growth is impaired in Hmga1a transgenic mice crossed on to an Mmp-2 deficient background. In human tumors, we discovered that HMGA1 is highest in aggressive carcinosarcomas and serous carcinomas, with lower levels in the more indolent endometrioid carcinomas. Moreover, HMGA1 and MMP-2 were positively correlated, but only in a subset of carcinosarcomas. HMGA1 also occupies the MMP-2 promoter in human carcinosarcoma cells. CONCLUSIONS Together, our studies define a novel HMGA1-MMP-2 pathway involved in a subset of human carcinosarcomas and tumor progression in murine models. Our work also suggests that targeting HMGA1 could be effective adjuvant therapy for more aggressive uterine cancers and provides compelling data for further preclinical studies.
Collapse
Affiliation(s)
- Joelle Hillion
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sujayita Roy
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mohammad Heydarian
- Department of Biologic Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Leslie Cope
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lingling Xian
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael Koo
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Li Z Luo
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathleen Kellyn
- Pathobiology Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brigitte M Ronnett
- Pathobiology Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tait Huso
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deborah Armstrong
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Karen Reddy
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biologic Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David L Huso
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - L M S Resar
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Pathobiology Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for Cellular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
24
|
Zeidler S, Meckbach C, Tacke R, Raad FS, Roa A, Uchida S, Zimmermann WH, Wingender E, Gültas M. Computational Detection of Stage-Specific Transcription Factor Clusters during Heart Development. Front Genet 2016; 7:33. [PMID: 27047536 PMCID: PMC4804722 DOI: 10.3389/fgene.2016.00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/23/2016] [Indexed: 12/28/2022] Open
Abstract
Transcription factors (TFs) regulate gene expression in living organisms. In higher organisms, TFs often interact in non-random combinations with each other to control gene transcription. Understanding the interactions is key to decipher mechanisms underlying tissue development. The aim of this study was to analyze co-occurring transcription factor binding sites (TFBSs) in a time series dataset from a new cell-culture model of human heart muscle development in order to identify common as well as specific co-occurring TFBS pairs in the promoter regions of regulated genes which can be essential to enhance cardiac tissue developmental processes. To this end, we separated available RNAseq dataset into five temporally defined groups: (i) mesoderm induction stage; (ii) early cardiac specification stage; (iii) late cardiac specification stage; (iv) early cardiac maturation stage; (v) late cardiac maturation stage, where each of these stages is characterized by unique differentially expressed genes (DEGs). To identify TFBS pairs for each stage, we applied the MatrixCatch algorithm, which is a successful method to deduce experimentally described TFBS pairs in the promoters of the DEGs. Although DEGs in each stage are distinct, our results show that the TFBS pair networks predicted by MatrixCatch for all stages are quite similar. Thus, we extend the results of MatrixCatch utilizing a Markov clustering algorithm (MCL) to perform network analysis. Using our extended approach, we are able to separate the TFBS pair networks in several clusters to highlight stage-specific co-occurences between TFBSs. Our approach has revealed clusters that are either common (NFAT or HMGIY clusters) or specific (SMAD or AP-1 clusters) for the individual stages. Several of these clusters are likely to play an important role during the cardiomyogenesis. Further, we have shown that the related TFs of TFBSs in the clusters indicate potential synergistic or antagonistic interactions to switch between different stages. Additionally, our results suggest that cardiomyogenesis follows the hourglass model which was already proven for Arabidopsis and some vertebrates. This investigation helps us to get a better understanding of how each stage of cardiomyogenesis is affected by different combination of TFs. Such knowledge may help to understand basic principles of stem cell differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Sebastian Zeidler
- University Medical Center Göttingen, Institute of Bioinformatics, Georg-August-University GöttingenGöttingen, Germany; Heart Research Center Göttingen, University Medical Center Göttingen, Institute of Pharmacology and Toxicology, Georg-August-University GöttingenGöttingen, Germany; DZHK (German Centre for Cardiovascular Research)Göttingen, Germany
| | - Cornelia Meckbach
- University Medical Center Göttingen, Institute of Bioinformatics, Georg-August-University Göttingen Göttingen, Germany
| | - Rebecca Tacke
- University Medical Center Göttingen, Institute of Bioinformatics, Georg-August-University Göttingen Göttingen, Germany
| | - Farah S Raad
- Heart Research Center Göttingen, University Medical Center Göttingen, Institute of Pharmacology and Toxicology, Georg-August-University GöttingenGöttingen, Germany; DZHK (German Centre for Cardiovascular Research)Göttingen, Germany
| | - Angelica Roa
- Heart Research Center Göttingen, University Medical Center Göttingen, Institute of Pharmacology and Toxicology, Georg-August-University GöttingenGöttingen, Germany; DZHK (German Centre for Cardiovascular Research)Göttingen, Germany
| | - Shizuka Uchida
- Institute of Cardiovascular Regeneration, Goethe University FrankfurtFrankfurt, Germany; DZHK (German Centre for Cardiovascular Research)Frankfurt, Germany
| | - Wolfram-Hubertus Zimmermann
- Heart Research Center Göttingen, University Medical Center Göttingen, Institute of Pharmacology and Toxicology, Georg-August-University GöttingenGöttingen, Germany; DZHK (German Centre for Cardiovascular Research)Göttingen, Germany
| | - Edgar Wingender
- University Medical Center Göttingen, Institute of Bioinformatics, Georg-August-University GöttingenGöttingen, Germany; DZHK (German Centre for Cardiovascular Research)Göttingen, Germany
| | - Mehmet Gültas
- University Medical Center Göttingen, Institute of Bioinformatics, Georg-August-University Göttingen Göttingen, Germany
| |
Collapse
|
25
|
Tian Y, Xu T, Huang J, Zhang L, Xu S, Xiong B, Wang Y, Tang H. Tissue Metabonomic Phenotyping for Diagnosis and Prognosis of Human Colorectal Cancer. Sci Rep 2016; 6:20790. [PMID: 26876567 PMCID: PMC4753490 DOI: 10.1038/srep20790] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/12/2016] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide and prognosis based on the conventional histological grading method for CRC remains poor. To better the situation, we analyzed the metabonomic signatures of 50 human CRC tissues and their adjacent non-involved tissues (ANIT) using high-resolution magic-angle spinning (HRMAS) (1)H NMR spectroscopy together with the fatty acid compositions of these tissues using GC-FID/MS. We showed that tissue metabolic phenotypes not only discriminated CRC tissues from ANIT, but also distinguished low-grade tumor tissues (stages I-II) from the high-grade ones (stages III-IV) with high sensitivity and specificity in both cases. Metabonomic phenotypes of CRC tissues differed significantly from that of ANIT in energy metabolism, membrane biosynthesis and degradations, osmotic regulations together with the metabolism of proteins and nucleotides. Amongst all CRC tissues, the stage I tumors exhibited largest differentiations from ANIT. The combination of the differentiating metabolites showed outstanding collective power for differentiating cancer from ANIT and for distinguishing CRC tissues at different stages. These findings revealed details in the typical metabonomic phenotypes associated with CRC tissues nondestructively and demonstrated tissue metabonomic phenotyping as an important molecular pathology tool for diagnosis and prognosis of cancerous solid tumors.
Collapse
Affiliation(s)
- Yuan Tian
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Tangpeng Xu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Jia Huang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Limin Zhang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shan Xu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bin Xiong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yulan Wang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310058, China
| | - Huiru Tang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Ministry of Education Key Laboratory of Contemporary Anthropology, Metabonomics and Systems Biology Laboratory, School of Life Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
26
|
Vartholomaiou E, Echeverría PC, Picard D. Unusual Suspects in the Twilight Zone Between the Hsp90 Interactome and Carcinogenesis. Adv Cancer Res 2015; 129:1-30. [PMID: 26915999 DOI: 10.1016/bs.acr.2015.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The molecular chaperone Hsp90 has attracted a lot of interest in cancer research ever since cancer cells were found to be more sensitive to Hsp90 inhibition than normal cells. Why that is has remained a matter of debate and is still unclear. In addition to increased Hsp90 dependence for some mutant cancer proteins and modifications of the Hsp90 machinery itself, a number of other characteristics of cancer cells probably contribute to this phenomenon; these include aneuploidy and overall increased numbers and levels of defective and mutant proteins, which all contribute to perturbed proteostasis. Work over the last two decades has demonstrated that many cancer-related proteins are Hsp90 clients, and yet only few of them have been extensively investigated, selected either on the basis of their obvious function as cancer drivers or because they proved to be convenient biomarkers for monitoring the effects of Hsp90 inhibitors. The purpose of our review is to go beyond these "usual suspects." We established a workflow to select poorly studied proteins that are related to cancer processes and qualify as Hsp90 clients. By discussing and taking a fresh look at these "unusual suspects," we hope to stimulate others to revisit them as novel therapeutic targets or diagnostic markers.
Collapse
Affiliation(s)
| | - Pablo C Echeverría
- Département de Biologie Cellulaire, Université de Genève, Sciences III, Geneva, Switzerland
| | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève, Sciences III, Geneva, Switzerland.
| |
Collapse
|
27
|
Williams MD, Zhang X, Park JJ, Siems WF, Gang DR, Resar LMS, Reeves R, Hill HH. Characterizing metabolic changes in human colorectal cancer. Anal Bioanal Chem 2015; 407:4581-95. [PMID: 25943258 DOI: 10.1007/s00216-015-8662-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 03/13/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death worldwide, despite the fact that it is a curable disease when diagnosed early. The development of new screening methods to aid in early diagnosis or identify precursor lesions at risk for progressing to CRC will be vital to improving the survival rate of individuals predisposed to CRC. Metabolomics is an advancing area that has recently seen numerous applications to the field of cancer research. Altered metabolism has been studied for many years as a means to understand and characterize cancer. However, further work is required to establish standard procedures and improve our ability to identify distinct metabolomic profiles that can be used to diagnose CRC or predict disease progression. The present study demonstrates the use of direct infusion traveling wave ion mobility mass spectrometry to distinguish metabolic profiles from CRC samples and matched non-neoplastic epithelium as well as metastatic and primary tumors at different stages of disease (T1-T4). By directly infusing our samples, the analysis time was reduced significantly, thus increasing the speed and efficiency of this method compared to traditional metabolomics platforms. Partial least squares discriminant analysis was used to visualize differences between the metabolic profiles of sample types and to identify the specific m/z features that led to this differentiation. Identification of the distinct m/z features was made using the human metabolome database. We discovered alterations in fatty acid biosynthesis and oxidative, glycolytic, and polyamine pathways that distinguish tumors from non-malignant colonic epithelium as well as various stages of CRC. Although further studies are needed, our results indicate that colonic epithelial cells undergo metabolic reprogramming during their evolution to CRC, and the distinct metabolites could serve as diagnostic tools or potential targets in therapy or primary prevention. Graphical Abstract Colon tissue biopsy samples were collected from patients after which metabolites were extracted via sonication. Two-dimensional data were collected via IMS in tandem with MS (IMMS). Data were then interpreted statistically via PLS-DA. Scores plots provided a visualization of statistical separation and groupings of sample types. Loading plots allowed identification of influential ion features. Lists of these features were exported and analyzed for specific differences. Direct comparisons of the ion features led to the identification and comparative analyses of candidate biomarkers. These differences were then expressed visually in charts and tables.
Collapse
Affiliation(s)
- Michael D Williams
- Department of Chemistry, Washington State University, Pullman, WA, 99164, USA
| | | | | | | | | | | | | | | |
Collapse
|