1
|
Cai R, Ke L, Zhao Y, Zhao J, Zhang H, Zheng P, Xin L, Ma C, Lin Y. NMR-based metabolomics combined with metabolic pathway analysis reveals metabolic heterogeneity of colorectal cancer tissue at different anatomical locations and stages. Int J Cancer 2025; 156:1644-1655. [PMID: 39629979 PMCID: PMC11826128 DOI: 10.1002/ijc.35273] [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: 05/25/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 02/16/2025]
Abstract
Colorectal cancer (CRC) still remains the leading cause of cancer death worldwide. This study aimed to profile the metabolic differences of colorectal cancer tissues (CCT) at different stages and sites, as compared with their distant noncancerous tissues (DNT), to investigate the temporal and spatial heterogeneities of metabolic characterization. Our NMR-based metabolomics fingerprinting revealed that many of the metabolite levels were significantly altered in CCT compared to DNT and esophageal cancer tissues, indicating deregulations of glucose metabolism, one-carbon metabolism, glutamine metabolism, amino acid metabolism, fatty acid metabolism, TCA cycle, choline metabolism, and so forth. A total of five biomarker metabolites, including glucose, glutamate, alanine, valine and histidine, were identified to distinguish between early and advanced stages of CCT. Metabolites that distinguish the different anatomical sites of CCT include glucose, glycerol, glutamine, inositol, succinate, and citrate. Those significant metabolic differences in CRC tissues at different pathological stages and sites suggested temporal and spatial heterogeneities of metabolic characterization in CCT, providing a metabolic foundation for further study on biofluid metabolism in CRC early detection.
Collapse
Affiliation(s)
- Rongzhi Cai
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - LiXin Ke
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - Yan Zhao
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - Jiayun Zhao
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - Huanian Zhang
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - Peie Zheng
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| | - Lijing Xin
- Animal Imaging and Technology Core, Center for Biomedical ImagingEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Changchun Ma
- Radiation Oncology DepartmentCancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Yan Lin
- Radiology Department, Second Affiliated HospitalShantou University Medical CollegeShantou CityGuangdong ProvinceChina
| |
Collapse
|
2
|
Chen Y, Zheng Z, Wang L, Chen R, He M, Zhao X, Jin L, Yao J. Deciphering STAT3's negative regulation of LHPP in ESCC progression through single-cell transcriptomics analysis. Mol Med 2024; 30:192. [PMID: 39468431 PMCID: PMC11520558 DOI: 10.1186/s10020-024-00962-0] [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: 06/15/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Esophageal Squamous Cell Carcinoma (ESCC) remains a predominant health concern in the world, characterized by high prevalence and mortality rates. Advances in single-cell transcriptomics have revolutionized cancer research by enabling a precise dissection of cellular and molecular diversity within tumors. OBJECTIVE This study aims to elucidate the cellular dynamics and molecular mechanisms in ESCC, focusing on the transcriptional influence of STAT3 (Signal Transducer and Activator of Transcription 3) and its interaction with LHPP, thereby uncovering potential therapeutic targets. METHODS Single-cell RNA sequencing was employed to analyze 44,206 cells from tumor and adjacent normal tissues of ESCC patients, identifying distinct cell types and their transcriptional shifts. We conducted differential gene expression analysis to assess changes within the tumor microenvironment (TME). Validation of key regulatory interactions was performed using qPCR in a cohort of 21 ESCC patients and further substantiated through experimental assays in ESCC cell lines. RESULTS The study revealed critical alterations in cell composition and gene expression across identified cell populations, with a notable shift towards pro-tumorigenic states. A significant regulatory influence of STAT3 on LHPP was discovered, establishing a novel aspect of ESCC pathogenesis. Elevated levels of STAT3 and suppressed LHPP expression were validated in clinical samples. Functional assays confirmed that STAT3 directly represses LHPP at the promoter level, and disruption of this interaction by promoter mutations diminished STAT3's repressive effect. CONCLUSION This investigation underscores the central role of STAT3 as a regulator in ESCC, directly impacting LHPP expression and suggesting a regulatory loop crucial for tumor behavior. The insights gained from our comprehensive cellular and molecular analysis offer a deeper understanding of the dynamics within the ESCC microenvironment. These findings pave the way for targeted therapeutic interventions focusing on the STAT3-LHPP axis, providing a strategic approach to improve ESCC management and prognosis.
Collapse
Affiliation(s)
- Yurao Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Zemao Zheng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Luoshai Wang
- Department of Thoracic Surgery, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
| | - Ronghuai Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Ming He
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Xiang Zhao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Liyan Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213000, Jiangsu, China.
- Department of Oncology, The Wujin Clinical college of Xuzhou Medical University, Changzhou, 213000, Jiangsu, China.
| | - Juan Yao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China.
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China.
| |
Collapse
|
3
|
Zhao Y, Liu H, Zhan Q, Jin H, Wang Y, Wang H, Huang B, Huang F, Jia X, Wang Y, Wang X. Oncolytic adenovirus encoding LHPP exerts potent antitumor effect in lung cancer. Sci Rep 2024; 14:13108. [PMID: 38849383 PMCID: PMC11161505 DOI: 10.1038/s41598-024-63325-z] [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: 01/22/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
LHPP has been shown to be a new tumor suppressor, and has a tendency to be under-expressed in a variety of cancers. Oncolytic virotheray is a promising therapeutics for lung cancer in recent decade years. Here we successfully constructed a new recombinant oncolytic adenovirus GD55-LHPP and investigated the effect of GD55-LHPP on the growth of lung cancer cells in vitro and in vivo. The results showed that LHPP had lower expression in either lung cancer cells or clinical lung cancer tissues compared with normal cells or tissues, and GD55-LHPP effectively mediated LHPP expression in lung cancer cells. GD55-LHPP could effectively inhibit the proliferation of lung cancer cell lines and rarely affected normal cell growth. Mechanically, the oncolytic adenovirus GD55-LHPP was able to induce stronger apoptosis of lung cancer cells compared with GD55 through the activation of caspase signal pathway. Notably, GD55-LHPP also activated autophagy-related signal pathway. Further, GD55-LHPP efficiently inhibited tumor growth in lung cancer xenograft in mice and prolonged animal survival rate compared with the control GD55 or PBS. In conclusion, the novel construct GD55-LHPP provides a valuable strategy for lung cancer-targeted therapy and develop the role of tumor suppress gene LHPP in lung cancer gene therapy.
Collapse
Affiliation(s)
- Yaru Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Huihui Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qi Zhan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Hao Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yiqiang Wang
- Surgical Department of Duchang County Second People's Hospital, Jiujiang, 332600, China
| | - Hui Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Xiaoyan Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China.
| |
Collapse
|
4
|
He R, Wang Y, Shuang C, Xu C, Li X, Cao Y. Single-cell transcriptomics reveals activation of endothelial cell and identifies LHPP as a potential target in ulcerative colitis. Heliyon 2024; 10:e29163. [PMID: 38601522 PMCID: PMC11004881 DOI: 10.1016/j.heliyon.2024.e29163] [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: 10/11/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
This study delves into Ulcerative colitis (UC), a persistent gastrointestinal disorder marked by inflammation and ulcers, significantly elevating colorectal cancer risk. The emergence of single-cell RNA sequencing (scRNA-seq) technology has opened new avenues for dissecting the intricate cellular dynamics and molecular mechanisms at play in UC pathology. By analyzing scRNA-seq data from individuals with UC, our study has revealed a consistent enhancement of inflammatory response pathways throughout the course of the disease, alongside detailing the characteristics of endothelial cell damage within colitis environments. A noteworthy finding is the downregulation of Phospholysine Phosphohistidine Inorganic Pyrophosphate Phosphatase (LHPP), which exhibited a inversely correlate with STAT3 expression levels. The markedly reduced expression of LHPP in both the tissues and plasma of UC patients positions LHPP as a compelling target for therapeutic intervention. Our findings highlight the pivotal role LHPP could play in moderating inflammation, spotlighting its potential as a crucial molecular target in the quest to understand and treat UC.
Collapse
Affiliation(s)
- Ruoyu He
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou Zhejiang, 310005, Zhejiang Province, China
| | - Yanfei Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou Zhejiang, 310005, Zhejiang Province, China
| | - Chen Shuang
- Department of Neurology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou, 310005, Zhejiang Province, China
| | - Chan Xu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou, 310005, Zhejiang Province, China
| | - Xiaoling Li
- Elder Medicine Department, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou, 310005, Zhejiang Province, China
| | - Yanfei Cao
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou Zhejiang, 310005, Zhejiang Province, China
| |
Collapse
|
5
|
Chen X, Xu Z, Zhou W, Zhang L, Huang J, Bao D. Single-Cell Transcriptomics Reveals STAT1 Drives LHPP Downregulation in Esophageal Squamous Cell Carcinoma Progression. Technol Cancer Res Treat 2024; 23:15330338241293485. [PMID: 39497541 PMCID: PMC11536641 DOI: 10.1177/15330338241293485] [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: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Esophageal Squamous Cell Carcinoma (ESCC) poses a significant health challenge in China due to its high incidence and mortality. Single-cell transcriptomics has transformed the approach to cancer research, allowing detailed analysis of cellular and molecular heterogeneity. The interaction between the transcription factor STAT1 and the tumor suppressor LHPP is crucial, potentially influencing cancer progression and therapeutic outcomes. OBJECTIVE This study aims to explore the molecular mechanisms and cellular dynamics underlying ESCC, with a focus on the role of transcription factors, particularly STAT1, and its regulatory relationship with LHPP, in the pathogenesis of ESCC. METHODS Utilizing single-cell transcriptomics data sourced from the public database GEO (GSE160269), we identified major cell types and their transcriptomic changes in ESCC patients. Differential gene expression profiles were examined to understand the dynamics of the tumor microenvironment (TME). A cohort of 21 ESCC patients was recruited to validate the findings. Furthermore, in ESCC cell lines, we validated the transcriptional regulatory relationship between STAT1 and LHPP. RESULTS Our analysis identified six major cell types within the ESCC microenvironment, revealing significant changes in cellular composition and gene expression profiles associated with tumorigenesis. Notably, a novel association between STAT1's regulatory role over LHPP was observed, suggesting a complex regulatory loop in ESCC pathogenesis. Elevated STAT1 and reduced LHPP expression were confirmed in patient samples with STAT1 negatively regulates LHPP expression at the promoter level; when the promoter binding motif regions were mutated, the transcriptional repression ability on LHPP was weakened. CONCLUSION The study highlights STAT1 as a core regulator in ESCC, directly influencing LHPP expression. The findings offer novel insights into the molecular mechanisms driving ESCC, shedding light on the cellular alterations and gene regulation dynamics within the ESCC microenvironment. This research provides a foundation for developing targeted therapeutic strategies, potentially utilizing the STAT1-LHPP axis as a focal point in ESCC treatment and prognosis.
Collapse
Affiliation(s)
- Xia Chen
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Zhiyun Xu
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Wubi Zhou
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Lili Zhang
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jian Huang
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Dawei Bao
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
6
|
Wang YF, He RY, Xu C, Li XL, Cao YF. Single-cell analysis identifies phospholysine phosphohistidine inorganic pyrophosphate phosphatase as a target in ulcerative colitis. World J Gastroenterol 2023; 29:6222-6234. [PMID: 38186864 PMCID: PMC10768394 DOI: 10.3748/wjg.v29.i48.6222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/27/2023] [Accepted: 12/14/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic gastrointestinal disorder characterized by inflammation and ulceration, representing a significant predisposition to colorectal cancer. Recent advances in single-cell RNA sequencing (scRNA-seq) technology offer a promising avenue for dissecting the complex cellular inter-actions and molecular signatures driving UC pathology. AIM To utilize scRNA-seq technology to dissect the complex cellular interactions and molecular signatures that underlie UC pathology. METHODS In this research, we integrated and analyzed the scRNA-seq data from UC patients. Moreover, we conducted mRNA and protein level assays as well as pathology-related staining tests on clinical patient samples. RESULTS In this study, we identified the sustained upregulation of inflammatory response pathways during UC progression, characterized the features of damaged endo-thelial cells in colitis. Furthermore, we uncovered the downregulation of phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) has a negative correlation with signal transducer and activator of transcription 3. Significant downregulation of LHPP in UC patient tissues and plasma suggests that LHPP may serve as a potential therapeutic target for UC. This paper highlights the importance of LHPP as a potential key target in UC and unveils its potential role in inflammation regulation. CONCLUSION The findings suggest that LHPP may serve as a potential therapeutic target for UC, emphasizing its importance as a potential key target in UC and unveiling its role in inflammation regulation.
Collapse
Affiliation(s)
- Yan-Fei Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou 310005, Zhejiang Province, China
| | - Ruo-Yu He
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou 310005, Zhejiang Province, China
| | - Chan Xu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou 310005, Zhejiang Province, China
| | - Xiao-Ling Li
- Elder Medicine Department, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou 310005, Zhejiang Province, China
| | - Yan-Fei Cao
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, No. 219 Moganshan Road, Xihu District, Hangzhou 310005, Zhejiang Province, China
| |
Collapse
|
7
|
Zhu H, Song C, Li J, Liu Q, Liu M, Fu L. LHPP suppresses proliferation, migration, and invasion in hepatocellular carcinoma and pancreatic cancer by inhibiting EGFR signaling pathway. Med Oncol 2023; 40:257. [PMID: 37522936 DOI: 10.1007/s12032-023-02127-1] [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: 05/03/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) has been reported to be a new tumor suppressor with a significant inhibitory effect in various cancers. Although LHPP has been repeatedly shown to inhibit the progression of various tumors by inhibiting the phosphorylation of AKT, up to now, the studies on the function and mechanism of LHPP in tumors are insufficient. In this study, LHPP expression was found to be downregulated in both hepatocellular carcinoma (HCC) and pancreatic cancer (PC). Here, we found that LHPP could bind to epidermal growth factor receptor (EGFR) and inhibit its phosphorylation, which thereby inhibited the activation of EGFR downstream pathways ERK, AKT, and STAT3, and then weakening the ability to proliferate, invade, and migrate in HCC and PC. This paper showed a new physiological function of LHPP in inhibiting phosphorylation of EGFR and its potential anti-tumor mechanism and indicated that LHPP was a potential therapeutic target for HCC and PC.
Collapse
Affiliation(s)
- Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China.
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China.
- Biobank, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China.
| | - Chunzhuo Song
- Guizhou Medical University, Guiyang, 550001, Guizhou, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
| | - Junjun Li
- Guizhou Medical University, Guiyang, 550001, Guizhou, China
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Qianfan Liu
- Guizhou Medical University, Guiyang, 550001, Guizhou, China
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, Jiangsu, China
| | - Meng Liu
- Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Liyue Fu
- Guizhou Medical University, Guiyang, 550001, Guizhou, China
| |
Collapse
|
8
|
Tumor suppressor LHPP suppresses cell proliferation and epithelial-mesenchymal transition in hepatocellular carcinoma cell lines. J Physiol Biochem 2022; 78:807-817. [PMID: 35796893 DOI: 10.1007/s13105-022-00903-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 06/03/2022] [Indexed: 10/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer in the world with high mortality due to its high potential of metastasis. Epithelial-mesenchymal transition (EMT) plays a key role in the pathogenesis of HCC occurrence and metastasis. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a novel tumor suppressor. There is little study about LHPP in human HCC development. In the present study, we aimed to investigate the role of LHPP in human HCC cell metastasis. We analyzed the LHPP expression level in human HCC tissues compared with normal tissues in the public database. We detected the mRNA level and protein level of LHPP in transformed liver cell line (LO2) and human HCC cell lines (MHCC-97 H, MHCC-97L, and HepG2). We performed genetic gain and loss of function experiments with LHPP using small interfering RNA (siRNA) and lentivirus infection. Then, we detected that LHPP suppressed proliferation and promoted apoptosis in hepatocellular carcinoma cell lines. Also, we investigated the role of LHPP in the EMT process. Finally, we examined the effect of LHPP on TGF-β-induced EMT. Interestingly, we also found that LHPP expression is positively regulated tumor suppressor p53. Our data showed that LHPP is significantly decreased in the human HCC tissues and human HCC cell lines compared with normal liver tissues and transformed liver cells. Knockdown of LHPP promotes HCC cell proliferation and metastasis, and LHPP expression levels negatively correlate with EMT-related genes. Furthermore, LHPP inhibits TGF-β-induced EMT in HCC cell lines. These studies validate LHPP as a tumor suppressor in liver cancer and provide a new genetic target for HCC diagnosis and treatment.
Collapse
|
9
|
Poursheikhani A, Abbaszadegan MR, Kerachian MA. Long non-coding RNA AC087388.1 as a novel biomarker in colorectal cancer. BMC Cancer 2022; 22:196. [PMID: 35193569 PMCID: PMC8862536 DOI: 10.1186/s12885-022-09282-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Background Several investigations have reported diverse roles of long non-coding RNA (lncRNA) in biological processes, tumor development, and progression of colorectal cancer (CRC). In this study, we investigated the lncRNA AC087388.1 tumorigenic role in CRC cells. Methods The CRC tissues were collected at the Reza Radiotherapy and Oncology Center, Mashhad, Iran. The human SW-48 and HT-29 CRC cell lines were obtained from the national cell bank of Iran. The cells were cultured according to ATCC (the American Type Culture Collection) recommendations. Quantitative real-time PCR was applied to assess the RNA expression. ShRNA transfection was done to downregulate the target gene. MTT and apoptosis assays were conducted to evaluate cell proliferation and viability, respectively. Colony formation assay, wound healing assay, and invasion assay were applied to determine growth, motility, and invasion of the cells, respectively. ENCORI online tool was used as downstream enrichment analysis. Results Forty CRC patients were encompassed in this study. The results demonstrated that the lncRNA SLC16A1-AS1, AC087388.1, and ELFN1-AS1 were significantly overexpressed in the CRC tissues in comparison to their normal counterpart margins. All the lncRNAs have shown significant Area Under Curve (AUC) values in the patients. Downregulation of lncRNA AC087388.1 remarkably decreased the cell proliferation and viability of the CRC cells. In addition, the data demonstrated that the downregulation of lncRNA AC087388.1 significantly suppressed cell growth and colony formation capability in the cells. Also, downregulation of lncRNA AC087388.1 attenuated motility and invasion of CRC cells, and significantly decreased the expression of invasion genes. In-silico functional enrichment analysis indicated that the lncRNA AC087388.1 has contributed to crucial signaling pathways in tumorigenesis such as the p53 and Wnt signaling pathways, apoptosis, and cell cycle. Conclusions Altogether, we showed that lncRNA AC087388.1 has an oncogenic role in tumorigenesis of CRC, and it can be considered as a novel diagnostic and prognostic biomarker in CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09282-0.
Collapse
Affiliation(s)
- Arash Poursheikhani
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Cancer Genetics Research Unit, Reza Radiotherapy and Oncology Center, Mashhad, Iran.
| |
Collapse
|
10
|
Guo C, Liu X, Xu Y, Han X, Xie R, Meng X, Li Y, Chen T, Cheng Z, Fu X. Exploring the Mechanism of Action of Canmei Formula Against Colorectal Adenoma Through Multi-Omics Technique. Front Cell Dev Biol 2021; 9:778826. [PMID: 34926462 PMCID: PMC8672438 DOI: 10.3389/fcell.2021.778826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Canmei formula (CMF) is a traditional Chinese medicine compound with definite effect on the prevention and treatment of colorectal adenoma (CRA). CMF can prevent the transformation of intestinal inflammation to cancer. This study explored the mechanism of action of CMF in anti-CRA using multi-omics techniques. Method: The mice were randomly divided into four groups: blank group (Control), high-fat diet (HFD) + AOM/DSS colorectal adenoma model (ADH) groups, Canmei formula treatment group (ADH-CMF) and sulfasalazine treatment group (Sul). Except for the blank group, ADH model was established in the other three groups by intraperitoneal injection with AOM reagent, and then mice were given 2.5% DSS in free drinking water and high-fat diet. The mice in the blank group and ADH groups were intragastrically perfused with normal saline, and the mice in the other two groups were treated with corresponding drugs for 20 weeks. During this period, the changes of physical signs of mice in each group were observed. The differentially expressed genes and proteins in the Control group, ADH group and ADH-CMF group were detected by RNA-seq transcriptome sequencing and Tandem Mass Tags (TMT) quantitative proteomics. After the combined analysis and verification, the key targets were analyzed by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Moreover, the changes of intestinal flora in mice of the three groups were examined. Results: A total of 2,548 differential genes were obtained by transcriptomics analysis, and 45 differential proteins were obtained by proteomics analysis. The results of proteomics data and experimental verification showed that CMF mainly affected the Phospholysine Phosphohistidine Inorganic Pyrophosphate Phosphatase (LHPP) target. GO analysis showed that the targets of CMF were involved in the biological processes such as cellular process, metabolic process and biological regulation. KEGG analysis showed that those genes were involved in oxidative phosphorylation, cell senescence, and metabolic pathways. Studies have shown that LHPP overexpression impeded colorectal cancer cell growth and proliferation in vitro, and was associated with a change in PI3K/AKT activity. The results of 16S DNA high-throughput sequencing showed that CMF could effectively regulate the abundance of Bifidobacterium, Candidatus_Saccharimonas and Erysipelatoclostridium in the intestinal flora at the genus level. Conclusion: CMF regulates LHPP via the PI3K/AKT signaling pathway. CMF affects the abundance of specific intestinal flora and can regulate the disorder of intestinal flora to achieve the role of prevention and treatment of CRA.
Collapse
Affiliation(s)
- Cui Guo
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Graduate School of Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Xiaoqiang Liu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Pain, Shibei Hospital, Shanghai, China
| | - Yimin Xu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Han
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Runnan Xie
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Miaohang Town Community Health Service Center, Shanghai, China
| | - Xiangxue Meng
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Graduate School of Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Yuan Li
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tongyu Chen
- Cardiothoracic Surgery Department, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhihong Cheng
- China State Institute of Pharmaceutical Industry, National Pharmaceutical Engineering Research Center, Shanghai, China
| | - Xiaoling Fu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
LHPP suppresses colorectal cancer cell migration and invasion in vitro and in vivo by inhibiting Smad3 phosphorylation in the TGF-β pathway. Cell Death Discov 2021; 7:273. [PMID: 34608127 PMCID: PMC8490460 DOI: 10.1038/s41420-021-00657-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
The roles of phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) in tumorigenesis have been recently proven in hepatocellular carcinoma (HCC), cervical, pancreatic, bladder, and thyroid cancers. Previous research demonstrated that LHPP repressed cell proliferation and growth by inactivating the phosphatidylinositol 3-kinase/AKT signaling pathway in vitro and in vivo. However, the functions and potential mechanisms of LHPP as a tumor suppressor in colorectal cancer (CRC) metastasis are still unknown. Consequently, the Transwell assay and xenograft nude model showed that LHPP inhibited migration and invasion of CRC cells in vitro and in vivo, respectively. The expression of total and nuclear epithelial-to-mesenchymal transition (EMT)-related proteins were significantly reduced after LHPP upregulation. Human Gene Expression Array and IPA (Ingenuity Pathway Analysis) commercial software were applied to identify differentially expressed genes (DEGs) and potential cell signaling pathways. A total of 330 different genes were observed, including 177 upregulated genes and 153 downregulated genes. Bioinformatics analysis suggested that the transforming growth factor-β (TGF-β) signaling pathway was highly inactivated in this study. Then, Smad3 phosphorylation was apparently decreased, whereas Smad7 expression was markedly enhanced after upregulating LHPP expression. These results were proven once again after TGF-β1 stimulation. Furthermore, a specific inhibitor of Smad3 phosphorylation (SIS3) was applied to verify that LHPP repressed EMT of cancer cells by attenuating TGF-β/Smad signaling. The results suggested that suppression of the TGF-β/Smad signaling pathway by LHPP overexpression could be abolished by SIS3.
Collapse
|
12
|
Guo C, Xu Y, Han X, Liu X, Xie R, Cheng Z, Fu X. Transcriptomic and Proteomic Study on the High-Fat Diet Combined With AOM/DSS-Induced Adenomatous Polyps in Mice. Front Oncol 2021; 11:736225. [PMID: 34513713 PMCID: PMC8427437 DOI: 10.3389/fonc.2021.736225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022] Open
Abstract
Objective To screen and identify molecular targets and bacteria genus leading to adenomatous polyps in mouse induced by high-fat diet (HFD) +AOM/DSS using omics technology. Methods The molecular targets of colorectal adenoma disease were obtained from the GeneCards and OMIM database. The SPF C57BL mice were randomly divided into blank (Control) and AOM/DSS+HFD colorectal adenoma model (ADH) groups. The ADH model group was intraperitoneally injected with AOM reagent. Then, mice were given with 2.5% DSS (in free drinking water) and high-fat diet to establish the mouse model. During this period, the changes of physical signs of mice in each group were observed. After the end of modeling, HE staining was used to evaluate the histopathological change of mice. The differentially expressed genes and proteins in the Control group and ADH group were detected by RNA-seq transcriptome sequencing and Tandem Mass Tags (TMT) quantitative proteomics. The histological results were analyzed by intersection with the intestinal adenoma molecular targets obtained from the database. Moreover, the changes of intestinal flora in the two groups were examined. The correlation between targets and differential bacteria was analyzed and verified by Parallel Reaction Monitoring (PRM) to comprehensively evaluate the mouse model of adenomatous polyp induced by AOM/DSS+HFD. Results The general condition and histopathological results of mice confirmed that the ADH mouse model was successfully established and tubular adenoma was formed. A total of 604 genes and 42 proteins related to intestinal adenoma were obtained by histological analysis and database intersection analysis. The intestinal microflora of ADH mice was different from that of normal mice, and the constituents and abundance of intestinal flora were similar to those of human intestinal adenoma. GATA4 and LHPP were selected as potential pathological markers of the model mice by correlation analysis of targets and intestinal flora. The results of PRM verification were highly consistent with the results of RNA-Seq transcriptome sequencing and TMT analysis. Conclusion The pathological results, molecular pathological markers and the changes of intestinal flora suggest that the mouse ADH model is ideal for studying the transformation of inflammatory cancer. The ADH model will be helpful for understanding the occurrence and development of human colorectal cancer at the transcriptomic and proteomic level.
Collapse
Affiliation(s)
- Cui Guo
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yimin Xu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Han
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoqiang Liu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Pain, Shibei Hospital, Shanghai, China
| | - Runnan Xie
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Traditional Chinese Medicine, Miaohang Town Community Health Service Center, Shanghai, China
| | - Zhihong Cheng
- China State Institute of Pharmaceutical Industry, National Pharmaceutical Engineering Research Center, Shanghai, China
| | - Xiaoling Fu
- Second Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Schinke CD, Bird JT, Qu P, Yaccoby S, Lyzogubov VV, Shelton R, Ling W, Boyle EM, Deshpande S, Byrum SD, Washam C, Mackintosh S, Stephens O, Thanendrarajan S, Zangari M, Shaughnessy J, Zhan F, Barlogie B, van Rhee F, Walker BA. PHF19 inhibition as a therapeutic target in multiple myeloma. Curr Res Transl Med 2021; 69:103290. [PMID: 33894670 DOI: 10.1016/j.retram.2021.103290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
Epigenetic deregulation is increasingly recognized as a contributing pathological factor in multiple myeloma (MM). In particular tri-methylation of H3 lysine 27 (H3K27me3), which is catalyzed by PHD finger protein 19 (PHF19), a subunit of the Polycomb Repressive Complex 2 (PRC2), has recently shown to be a crucial mediator of MM tumorigenicity. Overexpression of PHF19 in MM has been associated with worse clinical outcome. Yet, while there is mounting evidence that PHF19 overexpression plays a crucial role in MM carcinogenesis downstream mechanisms remain to be elucidated. In the current study we use a functional knock down (KD) of PHF19 to investigate the biological role of PHF19 and show that PHF19KD leads to decreased tumor growth in vitro and in vivo. Expression of major cancer players such as bcl2, myc and EGR1 were decreased upon PHF19KD further underscoring the role of PHF19 in MM biology. Additionally, our results highlighted the prognostic impact of PHF19 overexpression, which was significantly associated with worse survival. Overall, our study underscores the premise that targeting the PHF19-PRC2 complex would open up avenues for novel MM therapies.
Collapse
Affiliation(s)
- Carolina D Schinke
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Jordan T Bird
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Pingping Qu
- Cancer Research and Biostatistics, Seattle, WA, United States
| | - Shmuel Yaccoby
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Valeriy V Lyzogubov
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Randal Shelton
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Wen Ling
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Eileen M Boyle
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Sharyu Deshpande
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D Byrum
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Charity Washam
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Samuel Mackintosh
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Owen Stephens
- The College of Medicine, Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sharmilan Thanendrarajan
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Maurizio Zangari
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - John Shaughnessy
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Fenghuang Zhan
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Bart Barlogie
- Division of Hematology, The Mount Sinai Hospital, New York, NY, Sinai, USA
| | - Frits van Rhee
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Brian A Walker
- Division of Hematology Oncology, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
14
|
Chen GY, Chen JQ, Liu XY, Xu Y, Luo J, Wang YF, Zhou TL, Yan ZR, Zhou L, Tao QW. Total Flavonoids of Rhizoma Drynariae Restore the MMP/TIMP Balance in Models of Osteoarthritis by Inhibiting the Activation of the NF- κB and PI3K/AKT Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6634837. [PMID: 33995548 PMCID: PMC8081598 DOI: 10.1155/2021/6634837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/15/2021] [Accepted: 04/04/2021] [Indexed: 12/16/2022]
Abstract
Total flavonoids of Rhizoma Drynariae (TFRD) have been shown to have beneficial effects on osteoarthritis (OA) clinically, but the mechanisms have not been elucidated. In this study, we investigated the effect of TFRD on articular cartilage in an OA rat model established by the Hulth method and in SW1353 chondrocytes induced by the proinflammatory factor interleukin-1β (IL-1β). The results showed that TFRD could alleviate the pathological changes in knee cartilage in OA model rats. In vivo, the qPCR analysis indicated that the mRNA levels of matrix metalloproteinases, MMP-1, MMP-3, and MMP-13, were decreased, while tissue inhibitor of matrix metalloproteinases- (TIMP-) 4 was increased in cartilage, and these changes could be partially prevented by TFRD. In vitro experiments showed that IL-1β could significantly increase the expression of MMP-1, MMP-3, and MMP-13 and decrease the expression of TIMP-4 in SW1353 cells at the mRNA and protein levels. TFRD could increase the expression of MMP-3 and MMP-13 and decrease the expression of TIMP-4. Transfection of siRNA and addition of pathway inhibitors were used to clarify that inhibition of NF-κB and PI3K/AKT pathway decreased MMP-1, MMP-3, and MMP-13 and increased TIMP-4 expression. We also found that in IL-1β-induced SW1353 cells, TFRD pretreatment had a modest inhibitory effect on p-AKT (Ser473) and reversed the increase of nuclear factor kappa-B (NF-κB) p65 in nuclear fraction and the decrease of inhibitor of NF-κB(IκB)-α in the cytosolic fraction. Further immunofluorescence confirmed that TFRD can inhibit IL-1β-induced NF-κB p65 translocation to the nucleus to some extent. In conclusion, TFRD showed chondroprotective effects by restoring the MMP/TIMP balance in OA models by suppressing the activation of the NF-κB and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Guang-Yao Chen
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Qi Chen
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Yu Liu
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuan Xu
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing 100029, China
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jing Luo
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing 100029, China
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi-Fei Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tong-Liang Zhou
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ze-Ran Yan
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing 100029, China
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Li Zhou
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qing-Wen Tao
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing 100029, China
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
15
|
Chen WJ, Chen LH, Wang J, Wang ZT, Wu CY, Sun K, Ding BY, Liu N, Xu RX. LHPP impedes energy metabolism by inducing ubiquitin-mediated degradation of PKM2 in glioblastoma. Am J Cancer Res 2021; 11:1369-1390. [PMID: 33948363 PMCID: PMC8085851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/27/2021] [Indexed: 06/12/2023] Open
Abstract
Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a new-found tumor suppressor in a variety of tumors. While, it is still unknown about its role in glioma. In this study, we found that LHPP is abnormally decreasing or absent in glioblastoma, and the low expression of LHPP is associated with poor median survival in glioma patients. Functional assay revealed that LHPP-overexpression significantly inhibited U87MG and U118MG growth in vitro and in vivo. As to the mechanism, mass-spectrometric analysis indicated that the LHPP interacting proteins were mainly enriched in regulation of energy metabolism, including Carbon metabolism, Oxidative phosphorylation, and Glycolysis. Seahorse assay and metabolites detection confirmed that LHPP-overexpression obviously impeded glycolysis and respiration in U87MG and U118MG cells. For the further study, western blot assay showed that the protein level of PKM2 at dimeric, tetrameric, and total protein, were all decreased significantly, and its enzymatic activity was decreased as well. ChIP and RNAseq integrated analysis indicated that the decreased protein level of PKM2 was independent of PKM2 transcription, and LHPP did not reprogram transcription level of metabolic genome. Co-IP and immunofluorescence assay manifested that LHPP interacted with PKM2, and this interaction interfered the protein stability, then induced ubiquitin-mediated degradation of PKM2. Rescue assay confirmed that restoring the expression of PKM2 effectively reversed the restrained energy metabolism and the inhibited cancer cell growth caused by LHPP-overexpression in U87MG and U118MG cells. Taking together, we demonstrated that LHPP impedes the glycolysis and respiration during energy metabolic process via inducing ubiquitin-mediated degradation of PKM2, thus inhibits the growth of glioblastoma.
Collapse
Affiliation(s)
- Wen-Jin Chen
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510000, P. R. China
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| | - Li-Hua Chen
- The Department of Neurosurgery, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of ChinaChengdu 610072, P. R. China
| | - Ji Wang
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510000, P. R. China
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| | - Zhao-Tao Wang
- The Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510000, P. R. China
| | - Cui-Ying Wu
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| | - Kai Sun
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| | - Bo-Yun Ding
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510000, P. R. China
| | - Ning Liu
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| | - Ru-Xiang Xu
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510000, P. R. China
- Department of Neurosurgery, The Seventh Medical Center of General Hospital of PLABeijing 100010, P. R. China
| |
Collapse
|
16
|
Ding F, Yang S. Epigallocatechin-3-gallate inhibits proliferation and triggers apoptosis in colon cancer via the hedgehog/phosphoinositide 3-kinase pathways. Can J Physiol Pharmacol 2021; 99:910-920. [PMID: 33617370 DOI: 10.1139/cjpp-2020-0588] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The present study evaluated whether epigallocatechin-3-gallate (EGCG) effectively attenuates tumor growth in colon cancer cells and in the xenografts of nude mice and investigated the underlying mechanisms by focusing on the sonic hedgehog (Shh) and phosphoinositide 3-kinase (PI3K) pathways. Three kinds of colon cancer cells and BALB/c nude mice were used to evaluate the antiproliferative effect of EGCG. The apoptosis, migration, and invasion of colon cancer cells were analyzed to explore the toxicity effect of EGCG on colon cancer cells. Western blotting was used to demonstrate the expression levels of related proteins. The results showed that EGCG exhibited an antiproliferative effect against colon cancer cells in a dose-dependent manner with low toxicity against normal colon epithelial cells. Administration of EGCG caused significant apoptosis and inhibited the migration and invasion of colon cancer cells. The toxic effect of EGCG on colon cancer cells was accompanied by downregulation of the Shh and PI3K/Akt pathways. In addition, EGCG reduced tumor volume and weight without affecting the body weight of nude mice and inhibited the activation of the Shh and PI3K/AKT pathways in tumor tissue. Further study showed that purmorphamine (smoothened (Smo) agonist) or insulin like growth factor-1 (IGF-1, PI3K agonist) partly abolished the effect of EGCG on cell proliferation, migration, and apoptosis. Cyclopamine (Smo inhibitor) and LY294002 (PI3K inhibitor) showed the similar toxic effects as EGCG on colon cancer cells. In conclusion, EGCG inhibited colon tumor growth via downregulation of the Shh and PI3K pathways and may be a potential chemotherapeutic agent against colon cancer.
Collapse
Affiliation(s)
- Feng Ding
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Su Yang
- Department of Urology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
17
|
Qian J, Gong ZC, Zhang YN, Wu HH, Zhao J, Wang LT, Ye LJ, Liu D, Wang W, Kang X, Sheng J, Xu W, Liu XL, Wu J, Zheng W. Lactic acid promotes metastatic niche formation in bone metastasis of colorectal cancer. Cell Commun Signal 2021; 19:9. [PMID: 33478523 PMCID: PMC7818572 DOI: 10.1186/s12964-020-00667-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 09/22/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND To investigate the effect of lactic acid (LA) on the progression of bone metastasis from colorectal cancer (CRC) and its regulatory effects on primary CD115 (+) osteoclast (OC) precursors. METHODS The BrdU assay, Annexin-V/PI assay, TRAP staining and immunofluorescence were performed to explore the effect of LA on the proliferation, apoptosis and differentiation of OC precursors in vitro and in vivo. Flow cytometry was performed to sort primary osteoclast precursors and CD4(+) T cells and to analyze the change in the expression of target proteins in osteoclast precursors. A recruitment assay was used to test how LA and Cadhein-11 regulate the recruitment of OC precursors. RT-PCR and Western blotting were performed to analyze the changes in the mRNA and protein expression of genes related to the PI3K-AKT pathway and profibrotic genes. Safranin O-fast green staining, H&E staining and TRAP staining were performed to analyze the severity of bone resorption and accumulation of osteoclasts. RESULTS LA promoted the expression of CXCL10 and Cadherin-11 in CD115(+) precursors through the PI3K-AKT pathway. We found that CXCL10 and Cadherin-11 were regulated by the activation of CREB and mTOR, respectively. LA-induced overexpression of CXCL10 in CD115(+) precursors indirectly promoted the differentiation of osteoclast precursors through the recruitment of CD4(+) T cells, and the crosstalk between these two cells promoted bone resorption in bone metastasis from CRC. On the other hand, Cadherin-11 mediated the adhesion between osteoclast precursors and upregulated the production of specific collagens, especially Collagen 5, which facilitated fibrotic changes in the tumor microenvironment. Blockade of the PI3K-AKT pathway efficiently prevented the progression of bone metastasis caused by lactate. CONCLUSION LA promoted metastatic niche formation in the tumor microenvironment through the PI3K-AKT pathway. Our study provides new insight into the role of LA in the progression of bone metastasis from CRC. Video Abstract.
Collapse
Affiliation(s)
- Jin Qian
- College of Medicine, Southwest Jiaotong University, North Section 1 No.111, Second Ring Road, Chengdu, 610000 People’s Republic of China
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Zi-chen Gong
- College of Medicine, Southwest Jiaotong University, North Section 1 No.111, Second Ring Road, Chengdu, 610000 People’s Republic of China
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Yi-na Zhang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Hong-hua Wu
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Li-ting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Li-juan Ye
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Da Liu
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Wei Wang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Xia Kang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Jun Sheng
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Wei Xu
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Xi-lin Liu
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Juan Wu
- Department of Pharmacy, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Wei Zheng
- College of Medicine, Southwest Jiaotong University, North Section 1 No.111, Second Ring Road, Chengdu, 610000 People’s Republic of China
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| |
Collapse
|
18
|
Li C, Yang J, Wang W, Li R. LHPP exerts a tumor-inhibiting role in glioblastoma via the downregulation of Akt and Wnt/β-catenin signaling. J Bioenerg Biomembr 2021; 53:61-71. [PMID: 33394310 DOI: 10.1007/s10863-020-09866-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022]
Abstract
Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) has been recently identified as a novel inhibitor of multiple tumors; however, its role in glioblastoma (GBM) has not been investigated. This study aimed to evaluate whether LHPP exerts a potential tumor-inhibiting role in GBM. Compared with that in normal tissues, LHPP expression was lower in GBM tissues and various GBM cell lines. LHPP up-regulation in GBM cells markedly reduced their proliferation and invasion, and its knockdown had an oncogenic effect on these cells. Further studies revealed that overexpressed LHPP decreased the levels of Akt and glycogen synthase-3β phosphorylation and down-regulated Wnt/β-catenin signaling. By contrast, LHPP knockdown produced opposite effects. Akt suppression markedly abrogated the activation of Wnt/β-catenin signaling induced by LHPP knockdown. The reactivation of Wnt/β-catenin signaling partially reversed the inhibition of tumor growth in GBM mediated by LHPP overexpression. In addition, LHPP overexpression markedly retarded the tumorigenesis of GBM cells in vivo. These findings revealed that LHPP acts a potential inhibitor of tumor growth in GBM, and its overexpression represses GBM proliferation and invasion by down-regulating Akt and Wnt/β-catenin signaling. This work highlights the crucial role of LHPP in GBM progression and suggests its potential as an anticancer target for the treatment of this disease.
Collapse
Affiliation(s)
- Chuankun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
| | - Jingya Yang
- The Department of Operation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Ruichun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| |
Collapse
|
19
|
Adam K, Ning J, Reina J, Hunter T. NME/NM23/NDPK and Histidine Phosphorylation. Int J Mol Sci 2020; 21:E5848. [PMID: 32823988 PMCID: PMC7461546 DOI: 10.3390/ijms21165848] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
The NME (Non-metastatic) family members, also known as NDPKs (nucleoside diphosphate kinases), were originally identified and studied for their nucleoside diphosphate kinase activities. This family of kinases is extremely well conserved through evolution, being found in prokaryotes and eukaryotes, but also diverges enough to create a range of complexity, with homologous members having distinct functions in cells. In addition to nucleoside diphosphate kinase activity, some family members are reported to possess protein-histidine kinase activity, which, because of the lability of phosphohistidine, has been difficult to study due to the experimental challenges and lack of molecular tools. However, over the past few years, new methods to investigate this unstable modification and histidine kinase activity have been reported and scientific interest in this area is growing rapidly. This review presents a global overview of our current knowledge of the NME family and histidine phosphorylation, highlighting the underappreciated protein-histidine kinase activity of NME family members, specifically in human cells. In parallel, information about the structural and functional aspects of the NME family, and the knowns and unknowns of histidine kinase involvement in cell signaling are summarized.
Collapse
Affiliation(s)
| | | | | | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; (K.A.); (J.N.); (J.R.)
| |
Collapse
|
20
|
Zhang X, Kang H, Xiao J, Shi B, Li X, Chen G. LHPP Inhibits the Proliferation and Metastasis of Renal Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7020924. [PMID: 33426063 PMCID: PMC7772040 DOI: 10.1155/2020/7020924] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) is one of the ten most common cancers in the globe. Despite the diagnosis and treatment of renal cell carcinoma that have made great improvements, the morbidity and mortality rates of renal cell carcinoma remain unchanged remarkably. LHPP is a kind of histidine phosphatases, acting as a tumor suppressor in the progression of various cancers. In this study, we found that LHPP was significantly downregulated in RCC tissues and cell lines. Decreased expression of LHPP was closely correlated with tumor size and postoperative metastasis of RCC patients. In addition, overexpression of LHPP inhibited the proliferation and metastasis of RCC. However, suppression of LHPP promoted the proliferation and metastasis of RCC. In conclusion, our results presented the important role of LHPP in the development and progression of RCC.
Collapse
Affiliation(s)
- Xiaoting Zhang
- Shenzhen Bao'an District Songgang People's Hospital, Shenzhen 518100, China
| | - Huaning Kang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jing Xiao
- Shenzhen Bao'an District Songgang People's Hospital, Shenzhen 518100, China
| | - Benyan Shi
- Shenzhen Bao'an District Songgang People's Hospital, Shenzhen 518100, China
| | - Xiaofeng Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Guihong Chen
- Shenzhen Bao'an District Songgang People's Hospital, Shenzhen 518100, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|