1
|
Zhang S, Wang J, Chen Y, Liang W, Liu H, Du R, Sun Y, Hu C, Shang Z. CAFs-derived lactate enhances the cancer stemness through inhibiting the MST1 ubiquitination degradation in OSCC. Cell Biosci 2024; 14:144. [PMID: 39605072 PMCID: PMC11603751 DOI: 10.1186/s13578-024-01329-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), a predominant stromal cell type in the tumor microenvironment, significantly affect the progression of oral squamous cell carcinoma (OSCC). RESULTS The specific mechanisms through which CAFs influence the cancer stem cell phenotype in OSCC are not fully understood. This study explored the effects of lactic acid produced by CAFs on the cancer stem cells (CSCs) phenotype of OSCC cells. Our results demonstrated that CAFs exhibit increased glycolysis and lactic acid production. Lactic acid treatment enhances CSCs-related markers expression, sphere formation, and clonogenic ability of OSCC cells. RNA sequencing revealed that lactic acid treatment elevates Discs Large Homolog 5 (DLG5) expression and markedly affects the Hippo pathway. Further investigation revealed that DLG5 mediates the effects of lactic acid on the CSCs phenotype. DLG5 knockdown results in elevated expression of E3 ubiquitin ligase Cullin 3, which can promote the ubiquitination and degradation of MST1, but the expression of phosphorylated MST1 remains unchanged. This leads to enhanced binding of phosphorylated MST1 to YAP1, increasing YAP1 phosphorylation and activating the Hippo pathway. CONCLUSION Collectively, our findings suggest that lactic acid from CAFs promotes the CSCs phenotype in OSCC through the DLG5/CUL3/MST1 axis. Therefore, targeting lactic acid exchange between CAFs and tumor cells may provide a novel therapeutic approach to suppress the CSCs phenotype in OSCC.
Collapse
Affiliation(s)
- Shuzhen Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Central Department School & Hospital of Stomatology, Wuhan University, Wuhan, 430022, China
| | - Jingjing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Weilian Liang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ruixue Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yunqing Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
2
|
Huang Z, Li M, Gu B, Chen J, Liu S, Tan P, Fu W. Ferroptosis-related LINC02535/has-miR-30c-5p/EIF2S1 axis as a novel prognostic biomarker involved in immune infiltration and progression of PDAC. Cell Signal 2024; 123:111338. [PMID: 39117252 DOI: 10.1016/j.cellsig.2024.111338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND PDAC, also known as pancreatic ductal adenocarcinoma, is often diagnosed at a late stage due to nonspecific symptoms and a distinct lack of reliable biomarkers for timely diagnosis. Ferroptosis, a novel non-apoptotic cell death mode discovered in recent years, is strongly linked to the progression of PDAC and the evasion of the immune system. The objective of this study is to discover a novel ceRNA biomarker associated with ferroptosis and investigate its possible molecular mechanisms and therapeutic potential in PDAC. METHODS Based on the FerrDb and TCGA databases, the R survival package was used to screen for ferroptosis-related mRNAs associated with PDAC prognosis. The ferroptosis-related ceRNA network was identified by miRTarBase, miRNet, and starBase and visualized using Cytoscape. The LASSO regression analysis was used to build a risk model associated with ceRNA. Additionally, we investigated the correlation between the ceRNA axis and the infiltration of immune cells in PDAC by employing the ssGSEA algorithm. Spearman correlation analysis was used to investigate the association between the ceRNA network and the expression levels of immune checkpoint genes in PDAC. The prediction of potential medications for PAAD patients with high risk scores was conducted using the R package oncoPredict and the Genomics of Drug Sensitivity in Cancer (GDSC) repository. Expression levels of LINC02535 in clinical specimens and PDAC cell lines were determined using qRT-PCR. CCK-8, colony formation, EdU, wound healing, and transwell assays were performed to assess the impact of reducing LINC02535 on the growth, migration, and invasion of PDAC cell lines BxPC3 and PANC1. RESULTS We first discovered a new LINC02535/miR-30c-5p/EIF2S1 axis associated with ferroptosis and created a prognostic nomogram for predicting overall survival. Meanwhile, the risk scores of the LINC02535/miR-30c-5p/EIF2S1 axis associated with ferroptosis were linked to immune subtypes in PDAC. The high immune infiltration subtype exhibited elevated ceRNA risk scores and EIF2S1 expression. The correlation analysis revealed a positive correlation between ceRNA risk scores and four immune cells, namely Activated CD4 T cell, Memory B cell, Neutrophil, and Type 2 T helper cell, as well as four immune checkpoint genes, namely CD274, HAVCR2, PDCD1LG2, and TIGIT. The analysis of drug sensitivity indicated that individuals with a high-risk score may exhibit greater sensitivity to inhibitors targeting MEK1/2 compared to those with a low-risk score. In our validation experiments, it was observed that the expression of LINC02535 was increased in both PDAC tissues and cell lines. Additionally, the inhibition of LINC02535 resulted in decreased proliferation, migration, and invasion of PDAC cells. Rescue experiments demonstrated that LINC02535 promoted PDAC cell growth and metastasis by upregulating EIF2S1 expression. CONCLUSION To summarize, a novel ferroptosis-associated LINC02535/miR-30c-5p/EIF2S1 ceRNA network for PDAC patients was established. The analysis of this network's functionality offers potential insights for clinical decision-making and the advancement of precision medicine.
Collapse
MESH Headings
- Humans
- Ferroptosis/genetics
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Prognosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/immunology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Gene Expression Regulation, Neoplastic
- Disease Progression
- Cell Line, Tumor
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Mo Li
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Boyuan Gu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jiatong Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Shenglu Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Peng Tan
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
3
|
Li Z, Wang Y, Liang S, Yuan T, Liu J. EIF2S1 Silencing Impedes Neuroblastoma Development Through GPX4 Inactivation and Ferroptosis Induction. Int J Genomics 2024; 2024:6594426. [PMID: 39465005 PMCID: PMC11512646 DOI: 10.1155/2024/6594426] [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: 06/24/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Background: Neuroblastoma (NB) is one of the most devastating malignancies in children, accounting for a high mortality rate due to limited treatment options. This study is aimed at elucidating the role of the ferroptosis-related EIF2S1 gene in NB pathogenesis and exploring its potential as a therapeutic target. Methods: We conducted comprehensive bioinformatics analyses utilizing the FerrDb database and NB-related transcriptomics data to investigate the role of EIF2S1 in NB. Changes in EIF2S1 expression were subsequently validated in NB tissues and cell lines. Loss-of-function experiments were performed in SK-N-SH and IMR-32 cell lines through shRNA-mediated EIF2S1 knockdown. The impact of EIF2S1 knockdown on the tumorigenesis of SK-N-SH cells was assessed in nude mice. Results: Bioinformatics analyses revealed a significant association between elevated EIF2S1 expression and poor prognosis in NB patients. The increased levels of EIF2S1 expression were confirmed in NB tissues and cancerous cell lines. Furthermore, EIF2S1 overexpression was linked to translational regulation and immune cell infiltration modulation. Silencing of EIF2S1 resulted in the suppression of cell proliferation, migration, and tumorigenicity in NB cells. Additionally, EIF2S1 knockdown led to an accumulation of iron and oxidative stress, as well as a reduction in GPX4 and SLC7A11 expression. Conclusion: Our findings indicate that EIF2S1 appears to facilitate the progression of NB by protecting tumor cells from ferroptosis through modulating GPX4 and SLC7A11 expression. Consequently, EIF2S1 may serve as a potential therapeutic target for the management of NB.
Collapse
Affiliation(s)
- Zhen Li
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Yunhui Wang
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Shubin Liang
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Tingdong Yuan
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Jing Liu
- Department of Pathology, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| |
Collapse
|
4
|
Zhang Q, Ding L, Li J, Liu K, Xia C, Chen S, Huang X, Pu Y, Song Y, Hu Q, Wang Y. Single-cell RNA sequencing of OSCC primary tumors and lymph nodes reveals distinct origin and phenotype of fibroblasts. Cancer Lett 2024; 600:217180. [PMID: 39154702 DOI: 10.1016/j.canlet.2024.217180] [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: 03/04/2024] [Revised: 07/02/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Desmoplasia in fibroblasts within metastatic lymph nodes (MLNs) serves as an indicator of extranodal extension (ENE), which led mortality in oral squamous cell carcinoma (OSCC). However, systematic studies on fibroblasts in MLNs are lacking. Therefore, this study characterized the differences in phenotype, function, and origin of fibroblasts between primary tumors (PTs) and lymph nodes (LNs) in OSCC. We generated single-cell maps of PTs and paired MLNs and draining LNs from three OSCC patients. The transcriptomic atlas, pseudotime analysis, intercellular communication networks and enrichment analysis of the single cells were characterized. Phenotype and function heterogeneity of fibroblast cells between PTs and MLNs were further verified in vitro. Among 44,052 fibroblasts, we identified two distinct subpopulations of cancer-associated myofibroblastic cells (mCAFs): RGS4+ mCAF1 and COMP + mCAF2. Notably, they exhibited distinct distributions, with mCAF1 predominantly localized in the PTs and mCAF2 in the MLNs. Moreover, pseudotime analysis revealed their distinct origins: mCAF1 originated from inherent normal myofibroblastic cells in the PT, whereas mCAF2 originated from fibroblastic reticular cells in the LNs. Further functional experiments using primary fibroblasts revealed that, compared to mCAF1, mCAF2 in MLNs exhibited weaker crosstalk with immune cells but enhanced extracellular matrix activity, which is closely linked to ENE formation in OSCC. Additionally, we identified two fibroblast subgroups in a transforming state, indicating a potential epithelial-mesenchymal transition. Our research offers profound insights into the heterogeneity of fibroblasts between the PT and MLN in OSCC, serving as an essential resource for future drug discovery endeavors.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Jingyi Li
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Kunyu Liu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Chengwan Xia
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Sheng Chen
- Pathology Department, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Xiaofeng Huang
- Pathology Department, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Yumei Pu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China.
| | - Yuxin Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, China.
| |
Collapse
|
5
|
Zhang W, Zhang M, Ma J, Yao Y, Jiang Y, Huo Q, Jin S, Ji D, Zhao Y, Liu X, Sun H, Xu C, Zhang R. MicroRNA-15b promotes cardiac ischemia injury by the inhibition of Mitofusin 2/PERK pathway. Biochem Pharmacol 2024; 226:116372. [PMID: 38885773 DOI: 10.1016/j.bcp.2024.116372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/05/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
MicroRNA and mitofusin-2 (Mfn2) play an important role in the myocardial apoptosis induced by acute myocardial infarction (AMI). However, the target relationship and underlying mechanism associated with interorganelle interaction between endoplasmic reticulum (ER) and mitochondria under ischemic condition is not completely clear. MI-induced injury, Mfn2 expression, Mfn2-mediated mitochondrial function and ER stress, and target regulation by miRNA-15b (miR-15b) were evaluated by animal MI and cellular hypoxic models with advanced molecular techniques. The results confirmed that Mfn2 was down-regulated and miR-15b was up-regulated upon the target binding profile under ischemic/hypoxic condition. Our data showed that miR-15b caused cardiac apoptotic injury that was reversed by rAAV9-anti-miR-15b or AMO-15b. The damage effect of miR-15b on Mfn2 expression and mitochondrial function was observed and rescued by rAAV9-anti-miR-15b or AMO-15b. The targeted regulation of miR-15b on Mfn2 was verified by luciferase reporter and microRNA-masking. Importantly, miR-15b-mediated Mfn2 suppression activated PERK/CHOP pathway, by which leads to ER stress and mitochondrial dysfunction, and cardiac apoptosis eventually. In conclusion, our research, for the first time, revealed the missing molecular link in Mfn2 and apoptosis and elucidated that pro-apoptotic miR-15b plays crucial roles during the pathogenesis of AMI through down-regulation of Mfn2 and activation of PERK-mediated ER stress. These findings may provide an opportunity to develop new therapies for prophylaxis and treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Wenhao Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Mingyu Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jiao Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuan Yao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Qingji Huo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Saidi Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Dongni Ji
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yilin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinqi Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hao Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chaoqian Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Rong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
6
|
Zhang W, Shi Y, Oyang L, Cui S, Li S, Li J, Liu L, Li Y, Peng M, Tan S, Xia L, Lin J, Xu X, Wu N, Peng Q, Tang Y, Luo X, Liao Q, Jiang X, Zhou Y. Endoplasmic reticulum stress-a key guardian in cancer. Cell Death Discov 2024; 10:343. [PMID: 39080273 PMCID: PMC11289465 DOI: 10.1038/s41420-024-02110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) is a cellular stress response characterized by excessive contraction of the endoplasmic reticulum (ER). It is a pathological hallmark of many diseases, such as diabetes, obesity, and neurodegenerative diseases. In the unique growth characteristic and varied microenvironment of cancer, high levels of stress are necessary to maintain the rapid proliferation and metastasis of tumor cells. This process is closely related to ERS, which enhances the ability of tumor cells to adapt to unfavorable environments and promotes the malignant progression of cancer. In this paper, we review the roles and mechanisms of ERS in tumor cell proliferation, apoptosis, metastasis, angiogenesis, drug resistance, cellular metabolism, and immune response. We found that ERS can modulate tumor progression via the unfolded protein response (UPR) signaling of IRE1, PERK, and ATF6. Targeting the ERS may be a new strategy to attenuate the protective effects of ERS on cancer. This manuscript explores the potential of ERS-targeted therapies, detailing the mechanisms through which ERS influences cancer progression and highlighting experimental and clinical evidence supporting these strategies. Through this review, we aim to deepen our understanding of the role of ER stress in cancer development and provide new insights for cancer therapy.
Collapse
Grants
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 82302987, 82203233, 82202966, 82173142 National Natural Science Foundation of China (National Science Foundation of China)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- 2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2020JJ5336 Natural Science Foundation of Hunan Province (Hunan Provincial Natural Science Foundation)
- he Research Project of Health Commission of Hunan Province (202203034978, 202202055318, 202203231032, 202109031837, 202109032010, 20201020), Science and Technology Innovation Program of Hunan Province(2023ZJ1122, 2023RC3199, 2023RC1073), Hunan Provincial Science and Technology Department (2020TP1018), the Changsha Science and Technology Board (kh2201054), Ascend Foundation of National cancer center (NCC201909B06) and by Hunan Cancer Hospital Climb Plan (ZX2020001-3, YF2020002)
- the Research Project of Health Commission of Hunan Province (202203034978, 202202055318, 202203231032, 202109031837, 202109032010, 20201020), Science and Technology Innovation Program of Hunan Province(2023ZJ1122, 2023RC3199, 2023RC1073), Hunan Provincial Science and Technology Department (2020TP1018), the Changsha Science and Technology Board (kh2201054), Ascend Foundation of National cancer center (NCC201909B06) and by Hunan Cancer Hospital Climb Plan (ZX2020001-3, YF2020002)
Collapse
Affiliation(s)
- Wenlong Zhang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yidan Shi
- The High School Attached to Hunan Normal University, Changsha, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China
| | - Shiwen Cui
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shizhen Li
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinyun Li
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Lin Liu
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Yun Li
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Qianjin Liao
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China
- Department of Oncology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China.
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, China.
- Hunan Engineering Research Center of Tumor Organoids Technology and Application, Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, China.
| |
Collapse
|
7
|
Niu W, Wang B, Zhang Y, Wang C, Cao J, Li J, He Y, Lei P. Efficacy and safety evaluation of cross-reactive Fibroblast activation protein scFv-based CAR-T cells. Front Immunol 2024; 15:1433679. [PMID: 39086477 PMCID: PMC11288799 DOI: 10.3389/fimmu.2024.1433679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Fibroblast activation protein (FAP) overexpression on cancer-associated fibroblasts (CAFs) is associated with poor prognosis and worse clinical outcomes. Selective ablation of pro-tumorgenic FAP+ stromal cells with CAR-T cells may be a new therapeutic strategy. However, the clinical use of FAP-CAR T cells is suggested to proceed with caution for occasional poor efficacy and induction of on-target off-tumor toxicity (OTOT), including lethal osteotoxicity and cachexia. Hence, more investigations and preclinical trials are required to optimize the FAP-CAR T cells and to approve their safety and efficacy. Methods In this study, we designed second-generation CAR T cells targeting FAP with 4-1BB as a co-stimulatory molecule, and tested their cytotoxicity against FAP-positive cells (hFAP-HT1080 cells and a variety of primary CAFs) in vitro and in Cell line-derived xenograft (CDX) and a patient-derived xenograft (PDX) model. Results Results showed that our FAP-CAR T cells were powerfully potent in killing human and murine FAP-positive tumor cells and CAFs in multiple types of tumors in BALB/c and C57BL/6 mice and in patient-derived xenografts (PDX) model. And they were proved to be biologically safe and exhibit low-level OTOT. Discussion Taken together, the human/murine cross-reactive FAP-CAR T cells were powerfully potent in killing human and murine FAP positive tumor cells and CAFs. They were biologically safe and exhibit low-level OTOT, warranting further clinical investigation into our FAP-CAR T cells.
Collapse
Affiliation(s)
- Wenhao Niu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binchen Wang
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yirui Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaomin Wang
- Department of Clinical Laboratory, National Clinical Research Center for Cancer, Key laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jing Cao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Li
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yong He
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Yang M, Mu Y, Yu X, Gao D, Zhang W, Li Y, Liu J, Sun C, Zhuang J. Survival strategies: How tumor hypoxia microenvironment orchestrates angiogenesis. Biomed Pharmacother 2024; 176:116783. [PMID: 38796970 DOI: 10.1016/j.biopha.2024.116783] [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: 03/15/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
During tumor development, the tumor itself must continuously generate new blood vessels to meet their growth needs while also allowing for tumor invasion and metastasis. One of the most common features of tumors is hypoxia, which drives the process of tumor angiogenesis by regulating the tumor microenvironment, thus adversely affecting the prognosis of patients. In addition, to overcome unsuitable environments for growth, such as hypoxia, nutrient deficiency, hyperacidity, and immunosuppression, the tumor microenvironment (TME) coordinates angiogenesis in several ways to restore the supply of oxygen and nutrients and to remove metabolic wastes. A growing body of research suggests that tumor angiogenesis and hypoxia interact through a complex interplay of crosstalk, which is inextricably linked to the TME. Here, we review the TME's positive contribution to angiogenesis from an angiogenesis-centric perspective while considering the objective impact of hypoxic phenotypes and the status and limitations of current angiogenic therapies.
Collapse
Affiliation(s)
- Mengrui Yang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yufeng Mu
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Dandan Gao
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Jingyang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
9
|
Lu HJ, Koju N, Sheng R. Mammalian integrated stress responses in stressed organelles and their functions. Acta Pharmacol Sin 2024; 45:1095-1114. [PMID: 38267546 PMCID: PMC11130345 DOI: 10.1038/s41401-023-01225-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024]
Abstract
The integrated stress response (ISR) triggered in response to various cellular stress enables mammalian cells to effectively cope with diverse stressful conditions while maintaining their normal functions. Four kinases (PERK, PKR, GCN2, and HRI) of ISR regulate ISR signaling and intracellular protein translation via mediating the phosphorylation of eukaryotic translation initiation factor 2 α (eIF2α) at Ser51. Early ISR creates an opportunity for cells to repair themselves and restore homeostasis. This effect, however, is reversed in the late stages of ISR. Currently, some studies have shown the non-negligible impact of ISR on diseases such as ischemic diseases, cognitive impairment, metabolic syndrome, cancer, vanishing white matter, etc. Hence, artificial regulation of ISR and its signaling with ISR modulators becomes a promising therapeutic strategy for relieving disease symptoms and improving clinical outcomes. Here, we provide an overview of the essential mechanisms of ISR and describe the ISR-related pathways in organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosomes. Meanwhile, the regulatory effects of ISR modulators and their potential application in various diseases are also enumerated.
Collapse
Affiliation(s)
- Hao-Jun Lu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Nirmala Koju
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
10
|
Gu M, Liu Y, Xin P, Guo W, Zhao Z, Yang X, Ma R, Jiao T, Zheng W. Fundamental insights and molecular interactions in pancreatic cancer: Pathways to therapeutic approaches. Cancer Lett 2024; 588:216738. [PMID: 38401887 DOI: 10.1016/j.canlet.2024.216738] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The gastrointestinal tract can be affected by a number of diseases that pancreatic cancer (PC) is a malignant manifestation of them. The prognosis of PC patients is unfavorable and because of their diagnosis at advanced stage, the treatment of this tumor is problematic. Owing to low survival rate, there is much interest towards understanding the molecular profile of PC in an attempt in developing more effective therapeutics. The conventional therapeutics for PC include surgery, chemotherapy and radiotherapy as well as emerging immunotherapy. However, PC is still incurable and more effort should be performed. The molecular landscape of PC is an underlying factor involved in increase in progression of tumor cells. In the presence review, the newest advances in understanding the molecular and biological events in PC are discussed. The dysregulation of molecular pathways including AMPK, MAPK, STAT3, Wnt/β-catenin and non-coding RNA transcripts has been suggested as a factor in development of tumorigenesis in PC. Moreover, cell death mechanisms such as apoptosis, autophagy, ferroptosis and necroptosis demonstrate abnormal levels. The EMT and glycolysis in PC cells enhance to ensure their metastasis and proliferation. Furthermore, such abnormal changes have been used to develop corresponding pharmacological and nanotechnological therapeutics for PC.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
11
|
Guo Z, Ashrafizadeh M, Zhang W, Zou R, Sethi G, Zhang X. Molecular profile of metastasis, cell plasticity and EMT in pancreatic cancer: a pre-clinical connection to aggressiveness and drug resistance. Cancer Metastasis Rev 2024; 43:29-53. [PMID: 37453022 DOI: 10.1007/s10555-023-10125-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
The metastasis is a multistep process in which a small proportion of cancer cells are detached from the colony to enter into blood cells for obtaining a new place for metastasis and proliferation. The metastasis and cell plasticity are considered major causes of cancer-related deaths since they improve the malignancy of cancer cells and provide poor prognosis for patients. Furthermore, enhancement in the aggressiveness of cancer cells has been related to the development of drug resistance. Metastasis of pancreatic cancer (PC) cells has been considered one of the major causes of death in patients and their undesirable prognosis. PC is among the most malignant tumors of the gastrointestinal tract and in addition to lifestyle, smoking, and other factors, genomic changes play a key role in its progression. The stimulation of EMT in PC cells occurs as a result of changes in molecular interaction, and in addition to increasing metastasis, EMT participates in the development of chemoresistance. The epithelial, mesenchymal, and acinar cell plasticity can occur and determines the progression of PC. The major molecular pathways including STAT3, PTEN, PI3K/Akt, and Wnt participate in regulating the metastasis of PC cells. The communication in tumor microenvironment can provide by exosomes in determining PC metastasis. The components of tumor microenvironment including macrophages, neutrophils, and cancer-associated fibroblasts can modulate PC progression and the response of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Zhenli Guo
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, 128 Jinling Road, Ganzhou City, Jiangxi Province, 341000, China
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Gautam Sethi
- Department of Pharmacology, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
12
|
Maduabuchi WO, Tansi FL, Faenger B, Southern P, Pankhurst QA, Steiniger F, Westermann M, Hilger I. Local Magnetic Hyperthermia and Systemic Gemcitabine/Paclitaxel Chemotherapy Triggers Neo-Angiogenesis in Orthotopic Pancreatic Tumors without Involvement of Auto/Paracrine Tumor Cell VEGF Signaling and Hypoxia. Cancers (Basel) 2023; 16:33. [PMID: 38201461 PMCID: PMC10778317 DOI: 10.3390/cancers16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/20/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
There is a growing interest in exploring the therapeutically mediated modulation of tumor vascularization of pancreatic cancer, which is known for its poorly perfused tumor microenvironment limiting the delivery of therapeutic agents to the tumor site. Here, we assessed how magnetic hyperthermia in combination with chemotherapy selectively affects growth, the vascular compartment of tumors, and the presence of tumor cells expressing key regulators of angiogenesis. To that purpose, a orthotopic PANC-1 (fluorescent human pancreatic adenocarcinoma) mouse tumor model (Rj:Athym-Foxn1nu/nu) was used. Magnetic hyperthermia was applied alone or in combination with systemic chemotherapy (gemcitabine 50 mg per kg body weight, nab-pacitaxel 30 mg/kg body weight) on days 1 and 7 following magnetic nanoparticle application (dose: 1 mg per 100 mm3 of tumor). We used ultrasound imaging, immunohistochemistry, multi-spectral optoacoustic tomography (MSOT), and hematology to assess the biological parameters mentioned above. We found that magnetic hyperthermia in combination with gemcitabine/paclitaxel chemotherapy was able to impact tumor growth (decreased volumes and Ki67 expression) and to trigger neo-angiogenesis (increased small vessel diameter) as a result of the therapeutically mediated cell damages/stress in tumors. The applied stressors activated specific pro-angiogenic mechanisms, which differed from those seen in hypoxic conditions involving HIF-1α, since (a) treated tumors showed a significant decrease of cells expressing VEGF, CD31, HIF-1α, and neuropilin-1; and (b) the relative tumor blood volume and oxygen level remained unchanged. Neo-angiogenesis seems to be the result of the activation of cell stress pathways, like MAPK pathways (high number of pERK-expressing tumor cells). In the long term, the combination of magnetic hyperthermia and chemotherapy could potentially be applied to transiently modulate tumor angiogenesis and to improve drug accessibility during oncologic therapies of pancreatic cancer.
Collapse
Affiliation(s)
- Wisdom O. Maduabuchi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (F.L.T.); (B.F.)
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (F.L.T.); (B.F.)
| | - Bernd Faenger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (F.L.T.); (B.F.)
| | - Paul Southern
- Resonant Circuits Limited, 21 Albemarle Street, London W1S 4BS, UK; (P.S.); (Q.A.P.)
- Healthcare Biomagnetics Laboratory, University College London, 21 Albemarle St., London W1S 4BS, UK
| | - Quentin A. Pankhurst
- Resonant Circuits Limited, 21 Albemarle Street, London W1S 4BS, UK; (P.S.); (Q.A.P.)
- Healthcare Biomagnetics Laboratory, University College London, 21 Albemarle St., London W1S 4BS, UK
| | - Frank Steiniger
- Center for Electron Microscopy, Jena University Hospital, Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany; (F.S.); (M.W.)
| | - Martin Westermann
- Center for Electron Microscopy, Jena University Hospital, Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany; (F.S.); (M.W.)
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (F.L.T.); (B.F.)
| |
Collapse
|
13
|
Yin Z, Wang L. Endothelial-to-mesenchymal transition in tumour progression and its potential roles in tumour therapy. Ann Med 2023; 55:1058-1069. [PMID: 36908260 PMCID: PMC10795639 DOI: 10.1080/07853890.2023.2180155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
Tumour-associated endothelial cells (TECs) are a critical stromal cell type in the tumour microenvironment and play central roles in tumour angiogenesis. Notably, TECs have phenotypic plasticity, as they have the potential to transdifferentiate into cells with a mesenchymal phenotype through a process termed endothelial-to-mesenchymal transition (EndoMT). Many studies have reported that EndoMT influences multiple malignant biological properties of tumours, such as abnormal angiogenesis and tumour metabolism, growth, metastasis and therapeutic resistance. Thus, the value of targeting EndoMT in tumour treatment has received increased attention. In this review, we comprehensively explore the phenomenon of EndoMT in the tumour microenvironment and identify influencing factors and molecular mechanisms responsible for EndoMT induction. Furthermore, the pathological functions of EndoMT in tumour progression and potential therapeutic strategies for targeting EndoMT in tumour treatment are also discussed to highlight the pivotal roles of EndoMT in tumour progression and therapy.
Collapse
Affiliation(s)
- Zeli Yin
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liming Wang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
14
|
Yu ZZ, Xu BQ, Wang YY, Zhang PW, Shu YB, Shi Z. GSK2606414 Sensitizes ABCG2-Overexpressing Multidrug-Resistant Colorectal Cancer Cells to Chemotherapeutic Drugs. Biomedicines 2023; 11:3103. [PMID: 38002103 PMCID: PMC10669325 DOI: 10.3390/biomedicines11113103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Colorectal cancer is a common malignant tumor. A major factor in the high mortality rate of colorectal cancer is the emergence of multidrug resistance (MDR). Overexpression of the ABCG2 gene in cancer cells directly leads to MDR. Finding new inhibitors of ABCG2 may be an effective way to overcome drug resistance. We found that the compound GSK2606414 enhanced the sensitivity of the ABCG2 substrate to the chemotherapeutic drugs mitoxantrone and doxorubicin in ABCG2-overexpressing multidrug-resistant colorectal cancer cells by increasing their intracellular accumulation without affecting the protein expression of ABCG2. Molecular docking experiments predicted that GSK2606414 could stably bind in the drug-binding pocket of ABCG2. In conclusion, GSK2606414 can sensitize ABCG2-overexpressed multidrug-resistant colorectal cancer cells to chemotherapy drugs and can be used as a potential inhibitor of ABCG2.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Z.-Z.Y.); (B.-Q.X.); (Y.-Y.W.); (P.-W.Z.); (Y.-B.S.)
| |
Collapse
|
15
|
Monaco CF, Davis JS. Mechanisms of angioregression of the corpus luteum. Front Physiol 2023; 14:1254943. [PMID: 37841308 PMCID: PMC10568036 DOI: 10.3389/fphys.2023.1254943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
The corpus luteum is a transient ovarian endocrine gland that produces the progesterone necessary for the establishment and maintenance of pregnancy. The formation and function of this gland involves angiogenesis, establishing the tissue with a robust blood flow and vast microvasculature required to support production of progesterone. Every steroidogenic cell within the corpus luteum is in direct contact with a capillary, and disruption of angiogenesis impairs luteal development and function. At the end of a reproductive cycle, the corpus luteum ceases progesterone production and undergoes rapid structural regression into a nonfunctional corpus albicans in a process initiated and exacerbated by the luteolysin prostaglandin F2α (PGF2α). Structural regression is accompanied by complete regression of the luteal microvasculature in which endothelial cells die and are sloughed off into capillaries and lymphatic vessels. During luteal regression, changes in nitric oxide transiently increase blood flow, followed by a reduction in blood flow and progesterone secretion. Early luteal regression is marked by an increased production of cytokines and chemokines and influx of immune cells. Microvascular endothelial cells are sensitive to released factors during luteolysis, including thrombospondin, endothelin, and cytokines like tumor necrosis factor alpha (TNF) and transforming growth factor β 1 (TGFB1). Although PGF2α is known to be a vasoconstrictor, endothelial cells do not express receptors for PGF2α, therefore it is believed that the angioregression occurring during luteolysis is mediated by factors downstream of PGF2α signaling. Yet, the exact mechanisms responsible for angioregression in the corpus luteum remain unknown. This review describes the current knowledge on angioregression of the corpus luteum and the roles of vasoactive factors released during luteolysis on luteal vasculature and endothelial cells of the microvasculature.
Collapse
Affiliation(s)
- Corrine F. Monaco
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - John S. Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
- US Department of Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, NE, United States
| |
Collapse
|
16
|
Jia YZ, Liu J, Wang GQ, Pan H, Huang TZ, Liu R, Zhang Y. HIG1 domain family member 1A is a crucial regulator of disorders associated with hypoxia. Mitochondrion 2023; 69:171-182. [PMID: 36804467 DOI: 10.1016/j.mito.2023.02.009] [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] [Received: 06/08/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Mitochondria play a central role in cellular energy conversion, metabolism, and cell proliferation. The regulation of mitochondrial function by HIGD1A, which is located on the inner membrane of the mitochondria, is essential to maintain cell survival under hypoxic conditions. In recent years, there have been shown other cellular pathways and mechanisms involving HIGD1A diametrically or through its interaction. As a novel regulator, HIGD1A maintains mitochondrial integrity and enhances cell viability under hypoxic conditions, increasing cell resistance to hypoxia. HIGD1A mainly targets cytochrome c oxidase by regulating downstream signaling pathways, which affects the ATP generation system and subsequently alters mitochondrial respiratory function. In addition, HIGD1A plays a dual role in cell survival in distinct degree hypoxia regions of the tumor. Under mild and moderate anoxic areas, HIGD1A acts as a positive regulator to promote cell growth. However, HIGD1A plays a role in inhibiting cell growth but retaining cellular activity under severe anoxic areas. We speculate that HIGD1A engages in tumor recurrence and drug resistance mechanisms. This review will focus on data concerning how HIGD1A regulates cell viability under hypoxic conditions. Therefore, HIGD1A could be a potential therapeutic target for hypoxia-related diseases.
Collapse
Affiliation(s)
- Yin-Zhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Liu
- Key Laboratory of Coal Science and Technology of Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Geng-Qiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tie-Zeng Huang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Liu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
17
|
Naipauer J, Mesri EA. The Kaposi's sarcoma progenitor enigma: KSHV-induced MEndT-EndMT axis. Trends Mol Med 2023; 29:188-200. [PMID: 36635149 PMCID: PMC9957928 DOI: 10.1016/j.molmed.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023]
Abstract
Endothelial-to-mesenchymal transition has been described in tumors as a source of mesenchymal stroma, while the reverse process has been proposed in tumor vasculogenesis and angiogenesis. A human oncogenic virus, Kaposi's sarcoma herpes virus (KSHV), can regulate both processes in order to transit through this transition 'boulevard' when infecting KS oncogenic progenitor cells. Endothelial or mesenchymal circulating progenitor cells can serve as KS oncogenic progenitors recruited by inflammatory cytokines because KSHV can reprogram one into the other through endothelial-to-mesenchymal and mesenchymal-to-endothelial transitions. Through these novel insights, the identity of the potential oncogenic progenitor of KS is revealed while gaining knowledge of the biology of the mesenchymal-endothelial differentiation axis and pointing to this axis as a therapeutic target in KS.
Collapse
Affiliation(s)
- Julian Naipauer
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina; Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; University of Miami- Center for AIDS Research (UM-CFAR)/Sylvester Comprehensive Cancer Center (CCC) Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, FL, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Enrique A Mesri
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; University of Miami- Center for AIDS Research (UM-CFAR)/Sylvester Comprehensive Cancer Center (CCC) Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, FL, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
18
|
Correlation between hypoxia and HGF/c-MET expression in the management of pancreatic cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188869. [PMID: 36842767 DOI: 10.1016/j.bbcan.2023.188869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 02/28/2023]
Abstract
Pancreatic cancer (PC) is very deadly and difficult to treat. The presence of hypoxia has been shown to increase the probability of cancer developing and spreading. Pancreatic ductal adenocarcinoma (PDAC/PC) has traditionally viewed a highly lethal form of cancer due to its high occurrence of early metastases. Desmoplasia/stroma is often thick and collagenous, with pancreatic stellate cells as the primary source (PSCs). Cancer cells and other stromal cells interact with PSCs, promoting disease development. The hepatocyte growth factor (HGF)/c-MET pathway have been proposed as a growth factor mechanism mediating this interaction. Human growth factor (HGF) is secreted by pancreatic stellate cells (PSCs), and its receptor, c-MET, is generated by pancreatic cancer cells and endothelial cells. Hypoxia is frequent in malignant tumors, particularly pancreatic (PC). Hypoxia results from limitless tumor development and promotes survival, progression, and invasion. Hypoxic is becoming a critical driver and therapeutic target of pancreatic cancer as its hypoxia microenvironment is defined. Recent breakthroughs in cancer biology show that hypoxia promotes tumor proliferation, aggressiveness, and therapeutic resistance. Hypoxia-inducible factors (HIFs) stabilize hypoxia signaling. Hypoxia cMet is a key component of pancreatic tumor microenvironments, which also have a fibrotic response, that hypoxia, promotes and modulates. c-Met is a tyrosine-protein kinase. As describe it simply, the MET gene in humans' codes for a protein called hepatocyte growth factor receptor (HGFR). Most cancerous tumors and pancreatic cancer in particular, suffer from a lack of oxygen (PC). Due to unrestrained tumor development, hypoxia develops, actively contributing to tumor survival, progression, and invasion. As the processes by which hypoxia signaling promotes invasion and metastasis become clear, c-MET has emerged as an important determinant of pancreatic cancer malignancy and a potential pharmacological target. This manuscript provides the most current findings on the role of hypoxia and HGF/c-MET expression in the treatment of pancreatic cancer.
Collapse
|
19
|
Zhou Z, Saluja AK, Houchen CW, Li M. Replication stress identifies novel molecular classification associated with treatment outcomes in pancreatic cancer. Pancreatology 2023; 23:82-89. [PMID: 36435734 DOI: 10.1016/j.pan.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Replication stress is a prominent hallmark of tumor cells, which is crucial for maintaining genomic integrity. However, it remains poorly understood whether replication stress can serve as a surrogate biomarker to indicate prognosis and treatment response of pancreatic cancer. METHODS Transcriptomic and clinical data were obtained from The Cancer Genome Atlas and literature. An integrated signature of 18 replication-stress associated genes (termed as REST18) was established using the cox proportional hazards regression analysis. Tumors were sorted into REST18-low and REST18-high groups. Survival analysis, gene set enrichment analysis and composition of immune cells were compared between these tumors. RESULTS Patients with REST18-high tumors showed worse prognoses than those with REST18-low tumors in the TCGA database and the finding is validated in an independent cohort of pancreatic cancer. Comparison of REST18 model and other molecular classifications showed that REST18-high tumors are positively correlated to basal-like or squamous phenotypes, which have higher metastasis potential. DNA repair pathway is enriched in the REST18-high tumors. Analysis of tumor immune microenvironment found that REST18-high tumors are characterized with "immune-cold" features. Univariate and multivariate analysis show that REST18 is an independent risk factor for overall survival and predicts outcomes of chemotherapy in pancreatic cancer. CONCLUSION REST18 is a novel biomarker to indicate prognosis and treatment response of chemotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anuj K Saluja
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
20
|
Hypoxia activated HGF expression in pancreatic stellate cells confers resistance of pancreatic cancer cells to EGFR inhibition. EBioMedicine 2022; 86:104352. [PMID: 36371988 PMCID: PMC9664470 DOI: 10.1016/j.ebiom.2022.104352] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/18/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is an essential target for cancer treatment. However, EGFR inhibitor erlotinib showed limited clinical benefit in pancreatic cancer therapy. Here, we showed the underlying mechanism of tumor microenvironment suppressing the sensitivity of EGFR inhibitor through the pancreatic stellate cell (PSC). METHODS The expression of alpha-smooth muscle actin (α-SMA) and hypoxia marker in human pancreatic cancer tissues were detected by immunohistochemistry, and their correlation with overall survival was evaluated. Human immortalized PSC was constructed and used to investigate the potential effect on pancreatic cancer cell lines in hypoxia and normoxia. Luciferase reporter assay and Chromatin immunoprecipitation were performed to explore the potential mechanisms in vitro. The combined inhibition of EGFR and Met was evaluated in an orthotopic xenograft mouse model of pancreatic cancer. FINDINGS We found that high expression levels of α-SMA and hypoxia markers are associated with poor prognosis of pancreatic cancer patients. Mechanistically, we demonstrated that hypoxia induced the expression and secretion of HGF in PSC via transcription factor HIF-1α. PSC-derived HGF activates Met, the HGF receptor, suppressing the sensitivity of pancreatic cancer cells to EGFR inhibitor in a KRAS-independent manner by activating the PI3K-AKT pathway. Furthermore, we found that the combination of EGFR inhibitor and Met inhibitor significantly suppressed tumor growth in an orthotopic xenograft mouse model. INTERPRETATION Our study revealed a previously uncharacterized HIF1α-HGF-Met-PI3K-AKT signaling axis between PSC and cancer cells and indicated that EGFR inhibition plus Met inhibition might be a promising strategy for pancreatic cancer treatment. FUNDING This study was supported by The National Natural Science Foundation of China.
Collapse
|
21
|
Xu D, Liu Z, Liang MX, Fei YJ, Zhang W, Wu Y, Tang JH. Endoplasmic reticulum stress targeted therapy for breast cancer. Cell Commun Signal 2022; 20:174. [PMCID: PMC9639265 DOI: 10.1186/s12964-022-00964-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractRecurrence, metastasis, and drug resistance are still big challenges in breast cancer therapy. Internal and external stresses have been proven to substantially facilitate breast cancer progression through molecular and systemic mechanisms. For example, endoplasmic reticulum stress (ERS) results in activation of the unfolded protein response (UPR), which are considered an important cellular stress response. More and more reports indicate its key role in protein homeostasis and other diverse functions involved in the process of breast cancer progression. Therefore, therapies targeting the activation of ERS and its downstream signaling pathways are potentially helpful and novel tools to counteract and fight breast cancer. However, recent advances in our understanding of ERS are focused on characterizing and modulating ERS between healthy and disease states, and so little attention has been paid to studying the role and clinical application of targeting ERS in a certain cancer. In this review, we summarize the function and main mechanisms of ERS in different molecular types of breast cancer, and focus on the development of agents targeting ERS to provide new treatment strategies for breast cancer.
Collapse
|
22
|
Liu Y, Liang X, Zhang H, Dong J, Zhang Y, Wang J, Li C, Xin X, Li Y. ER Stress–Related Genes EIF2AK3, HSPA5, and DDIT3 Polymorphisms are Associated With Risk of Lung Cancer. Front Genet 2022; 13:938787. [PMID: 35923704 PMCID: PMC9341132 DOI: 10.3389/fgene.2022.938787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: This study aimed to evaluate the associations between endoplasmic reticulum (ER) stress–related genes EIF2AK3/PERK, HSPA5/GRP78, and DDIT3/CHOP polymorphisms and the risk of lung cancer. Methods: Six single-nucleotide polymorphisms (SNPs) of EIF2AK3, HSPA5, and DDIT3 were genotyped in 620 cases and 620 controls using a MassARRAY platform. Results: The minor allele A of rs6750998 was a protective allele against the risk of lung cancer (p < 0.001), while the minor alleles of rs867529, rs391957, and rs697221 were all risk alleles that may lead to multiplied risk of the disease (rprs867529 = 0.002; prs391957 = 0.015; prs697221 < 0.001). Moreover, the rs6750998-TA/AA genotypes were protective genotypes against the risk of lung cancer (p = 0.005); however, the rs867529-GC/CC, rs391957-CC, and rs697221-GA/AA genotypes were associated with elevated lung cancer risk (prs867529 = 0.003, prs391957 = 0.028, and prs697221 = 0.0001). In addition, EIF2AK3-rs6750998 was associated with a decreased risk of lung cancer under dominant, recessive, and log-additive models (p < 0.05). By contrast, the EIF2AK3-rs867529 was correlated with an increased risk of the disease under dominant and log-additive models (p = 0.001). Moreover, HSPA5-rs391957 was related to an elevated risk of the disease under recessive and log-additive models (p < 0.02). DDIT3-rs697221 was identified to have a significant association with the risk of lung cancer under all three genetic models (p < 0.01). Conclusion: Our results provide new insights on the role of the ER stress–related genes EIF2AK3, HSPA5, and DDIT3 polymorphisms for lung cancer risk.
Collapse
Affiliation(s)
- Yongshi Liu
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiaohua Liang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Hongpei Zhang
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jiajia Dong
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yan Zhang
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Juan Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Chunmei Li
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiangbing Xin
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiangbing Xin, ; Yan Li,
| | - Yan Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiangbing Xin, ; Yan Li,
| |
Collapse
|