1
|
Kim D, Allen CA, Chung D, Meng L, Zhang X, Zhang W, Ouyang Y, Li Z, Hong F. A novel TLR4 accessory molecule drives hepatic oncogenesis through tumor-associated macrophages. Cancer Lett 2025; 614:217543. [PMID: 39929433 DOI: 10.1016/j.canlet.2025.217543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment, yet the roles and mechanisms of TAMs in inflammation-associated oncogenesis remain enigmatic. We report that protein canopy homolog 2 (CNPY2) functions as a novel TLR4 regulator, promoting cytokine production in macrophages. CNPY2 binds directly to TLR4. Cnpy2 deficiency reduces cell surface expression of TLR4, nuclear translocation of NFκB and cytokine production in macrophages. Macrophage-specific CNPY2 deficiency significantly decreases cytokine production in macrophages and reduces hepatocarcinogenesis in a diethylnitrosamine (DEN)-induced liver cancer model. RNA-sequencing analysis revealed Cnpy2 knockout decreased the mRNA level and cell surface expression of two VEGF receptors, Flt1 and Kdr, compared to those in WT counterparts, resulting in inhibition of macrophage tumor infiltration. Cnpy2 knockout inhibits NFκB2/p52-mediated transcription of Flt1 and Kdr in macrophages. These findings demonstrate that CNPY2 regulates macrophages in both inflammation and hepatocarcinogenesis and may serve as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Doyeon Kim
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Carter A Allen
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Dongjun Chung
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Biostatistics Core, College of Nursing, College of Public Health, University of South Florida Health, 12901 Bruce B. Downs Blvd.Tampa, FL, 33612, USA
| | - Wenqing Zhang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Yuli Ouyang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Zihai Li
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Mullick Chowdhury S, Hong F, Rolfo C, Li Z, He K, Wesolowski R, Mortazavi A, Meng L. CNPY2 in Solid Tumors: Mechanisms, Biomarker Potential, and Therapeutic Implications. BIOLOGY 2025; 14:214. [PMID: 40001982 PMCID: PMC11851889 DOI: 10.3390/biology14020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Canopy FGF signaling regulator 2 (CNPY2) has emerged as a crucial player in cancer development by promoting cell proliferation, tissue repair, and angiogenesis. This review synthesizes the current understanding of CNPY2's role in solid tumors, particularly renal cell carcinoma, prostate cancer, hepatocellular carcinoma, and non-small-cell lung cancer. CNPY2 modulates key pathways such as p53, MYLIP, NF-κB, and AKT/GSK3β, thereby driving tumor growth and progression. In renal cell carcinoma, CNPY2 paradoxically promotes tumor growth through p53 upregulation, while in hepatocellular carcinoma, CNPY2 drives cell cycle progression via p53 destabilization. In prostate cancer, it enhances tumor progression by stabilizing androgen receptors through MYLIP interaction, and in non-small-cell lung cancer, it contributes to chemoresistance and metastasis through NF-κB and AKT/GSK3β signaling. Additionally, CNPY2 influences the tumor microenvironment, impacting immune function and metastatic potential. As a potential biomarker, CNPY2 shows promise for cancer detection and prognosis, particularly when used in combination with other markers. Early therapeutic strategies, including siRNA and miRNA approaches, are under exploration, though challenges remain due to CNPY2's expression in normal tissues and potential off-target effects. This review underscores the need for further research to fully elucidate CNPY2's oncogenic mechanisms and develop targeted therapies. Improved understanding of CNPY2's diverse roles may lead to novel diagnostic and therapeutic approaches in solid tumors.
Collapse
Affiliation(s)
- Sayan Mullick Chowdhury
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Feng Hong
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Christian Rolfo
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kai He
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| |
Collapse
|
3
|
Li X, Yin MY, Zhang ST, Xie SA. The role of canopy family proteins: biological mechanism and disease function. Mol Biol Rep 2025; 52:164. [PMID: 39869231 DOI: 10.1007/s11033-025-10269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
Canopy family proteins are highly sequence-conserved proteins with an N-terminal hydrophobic signal sequence, a unique pattern of six cysteine residues characteristic of the saposin-like proteins, and a C-terminal putative endoplasmic reticulum retention signal sequence. At present, the known canopy family proteins are canopy fibroblast growth factor signaling regulator 1 (CNPY1), CNPY2, CNPY3, and CNPY4. Despite similar structures, canopy family proteins regulate complex signal networks to participate in various biological processes. They are involved in a wide range of diseases, including angiogenesis, abnormal immune responses, neurodevelopmental disorders, and the development of tumors. Here, we summarized the biological processes and influence on the disease of every CNPY family protein to elucidate potential biomarkers and point out the direction for future in-depth research.
Collapse
Affiliation(s)
- Xue Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Min-Yue Yin
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Shu-Tian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Road, Xi Cheng District, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, State Key Laboratory of Digestive Health, Beijing, 100050, China.
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Road, Xi Cheng District, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, State Key Laboratory of Digestive Health, Beijing, 100050, China.
| |
Collapse
|
4
|
Wang JB, Ding SL, Liu XS, Yu T, Wu ZA, Li YX. Hypoxia Affects Mitochondrial Stress and Facilitates Tumor Metastasis of Colorectal Cancer Through Slug SUMOylation. Curr Mol Med 2025; 25:27-36. [PMID: 38013443 DOI: 10.2174/0115665240271525231112121008] [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: 07/11/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor. Slug has been found to display a key role in diversified cancers, but its relevant regulatory mechanisms in CRC development are not fully explored. OBJECTIVE Hence, exploring the function and regulatory mechanisms of Slug is critical for the treatment of CRC. METHODS Protein expressions of Slug, N-cadherin, E-cadherin, Snail, HIF-1α, SUMO- 1, Drp1, Opa1, Mfn1/2, PGC-1α, NRF1, and TFAM were measured through western blot. To evaluate the protein expression of Slug and SUMO-1, an immunofluorescence assay was used. Cell migration ability was tested through transwell assay. The SUMOylation of Slug was examined through CO-IP assay. RESULTS Slug displayed higher expression and facilitated tumor metastasis in CRC. In addition, hypoxia treatment was discovered to upregulate HIF-1α, Slug, and SUMO-1 levels, as well as induce Slug SUMOylation. Slug SUMOylation markedly affected mitochondrial biosynthesis, fusion, and mitogen-related protein expression levels to trigger mitochondrial stress. Additionally, the induced mitochondrial stress by hypoxia could be rescued by Slug inhibition and TAK-981 treatment. CONCLUSION Our study expounded that hypoxia affects mitochondrial stress and facilitates tumor metastasis of CRC through Slug SUMOylation.
Collapse
Affiliation(s)
- Jin-Bao Wang
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Shi-Lin Ding
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Xiao-Song Liu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Tianren Yu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Zeng-An Wu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Yu-Xiang Li
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| |
Collapse
|
5
|
Qiao Z, Li Y, Li S, Liu S, Cheng Y. Hypoxia-induced SHMT2 protein lactylation facilitates glycolysis and stemness of esophageal cancer cells. Mol Cell Biochem 2024; 479:3063-3076. [PMID: 38175377 DOI: 10.1007/s11010-023-04913-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024]
Abstract
Esophageal cancer (EC) is a familiar digestive tract tumor with highly lethal. The hypoxic environment has been demonstrated to be a significant factor in modulating malignant tumor progression and is strongly associated with the abnormal energy metabolism of tumor cells. Serine hydroxymethyl transferase 2 (SHMT2) is one of the most frequently expressed metabolic enzymes in human malignancies. The study was designed to investigate the biological functions and regulation mechanisms of SHMT2 in EC under hypoxia. We conducted RT-qPCR to assess SHMT2 levels in EC tissues and cells (TE-1 and EC109). EC cells were incubated under normoxia and hypoxia, respectively, and altered SHMT2 expression was evaluated through RT-qPCR, western blot, and immunofluorescence. The biological functions of SHMT2 on EC cells were monitored by performing CCK-8, EdU, transwell, sphere formation, glucose uptake, and lactate production assays. The SHMT2 protein lactylation was measured by immunoprecipitation and western blot. In addition, SHMT2-interacting proteins were analyzed by bioinformatics and validated by rescue experiments. SHMT2 was notably upregulated in EC tissues and cells. Hypoxia elevated SHMT2 protein expression, augmenting EC cell proliferation, migration, invasion, stemness, and glycolysis. In addition, hypoxia triggered lactylation of the SHMT2 protein and enhanced its stability. SHMT2 knockdown impeded the malignant phenotype of EC cells. Further mechanistic studies disclosed that SHMT2 is involved in EC progression by interacting with MTHFD1L. Hypoxia-induced SHMT2 protein lactylation and upregulated its protein level, which in turn enhanced MTHFD1L expression and accelerated the malignant progression of EC cells.
Collapse
Affiliation(s)
- Zhe Qiao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Yu Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Shaomin Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Shiyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Yao Cheng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
6
|
Wang L, Wang Y, Yang K, Hu X, Ye G. Roles of microRNA-486-5p in the diagnosis and the association with clinical symptoms of cervical cancer. Biomark Med 2024; 18:869-876. [PMID: 39417317 PMCID: PMC11508949 DOI: 10.1080/17520363.2024.2400963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024] Open
Abstract
Aim: To explore the predictive value of miR-486-5p in early cervical cancer and the associations of miR-486-5p with different clinical symptoms.Materials & methods: A total of 185 women were recruited. The relative expression levels of serum miR-486-5p were analyzed by real-time polymerase chain reaction. The receiver operator characteristic curves were utilized to reflect the predictive performance of miR-486-5p and squamous cell carcinoma antigen for early cervical cancer. Univariate logistic regression and ranked logistic regression were used to explore the associations of miR-486-5p with different clinical symptoms of early cervical cancer, with odd ratios (ORs) and 95% confidence intervals.Results: Eighty-one women (44.26%) had early cervical cancer. The relative expression of serum miR-486-5p was 1.99-fold higher in early cervical cancer patients than that in controls (p < 0.0001). The predictive performance of miR-486-5p for early cervical cancer was significantly superior to that of squamous cell carcinoma antigen, with an area under the curve of 0.865 (p < 0.001), sensitivity of 1.000 and specificity of 0.804. In addition, overexpressed miR-486-5p was associated with high odds of maximum tumor diameter increase (OR = 1.30, 95% CI: [1.01-1.66]).Conclusion: MiR-486-5p may be a potential biomarker for the early cervical cancer diagnosis, and was linked to the risk of maximum tumor diameter.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Gynecology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, PR China
| | - Yan Wang
- Department of Gynecology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, PR China
| | - Kang Yang
- Department of Gynecology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, PR China
| | - Xiaowen Hu
- Department of Gynecology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, PR China
| | - Guoliu Ye
- Department of Gynecology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, PR China
| |
Collapse
|
7
|
Yang W, An L, Li Y, Qian S. A cellular senescence-related genes model allows for prognosis and treatment stratification of cervical cancer: a bioinformatics analysis and external verification. Aging (Albany NY) 2023; 15:9408-9425. [PMID: 37768206 PMCID: PMC10564413 DOI: 10.18632/aging.204981] [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: 05/17/2023] [Accepted: 07/20/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Cervical cancer (CC) is highly lethal and aggressive with an increasing trend of mortality for females. Molecular characterization-based methods hold great promise for improving the diagnostic accuracy and for predicting treatment response. METHODS The mRNAs expression data of CC patients and cellular senescence-related genes were obtained from the Cancer Genome Atlas (TCGA) and CellAge databases, respectively. Differentially expressed genes (DEGs) of senescence related genes between tumor and normal tissues were used for Least absolute shrinkage and selection operator (LASSO) regression to construct a prognostic model. Univariate and LASSO regression analyses were applied to establish a predictive nomogram. The performance of the nomogram were evaluated by Kaplan-Meier curve, receiver operating characteristic (ROC), Harrell's concordance index (C-index), and calibration curve. GSE44001 and GSE52903 were used for external validation. RESULTS We established a cellular senescence-related genes-based stratified model, and a multivariable-based nomogram, which could accurately predict the prognosis of CC patients in the TCGA database. The Kaplan-Meier curve indicated that patients in the low-risk group had considerably better overall survival (OS, P =2.021e-05). The area under the ROC curve (AUC) of this model was 0.743 for OS. Multivariate analysis found that the 6-gene risk signature (HR=3.166, 95%CI: 1.660-6.041, P<0.001) was an independent risk factor for CC patients. We then designed an OS-associated nomogram that included the risk signature and clinicopathological factors. The AUC reached 0.860 for predicting 5-year OS. The nomogram showed excellent consistency between the predictions and actual survival observations. Two external GEO validations were corresponding to the gene expression pattern in TCGA. CONCLUSIONS Our results suggested a six-senescence related signature and established a prognostic nomogram that reliably predicted the overall survival for CC. These findings may be beneficial to personalized treatment and medical decision-making.
Collapse
Affiliation(s)
- Weiwei Yang
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Lijuan An
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Yanfei Li
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Sumin Qian
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| |
Collapse
|
8
|
Chen Y, Yan H, Yan L, Wang X, Che X, Hou K, Yang Y, Li X, Li Y, Zhang Y, Hu X. Hypoxia-induced ALDH3A1 promotes the proliferation of non-small-cell lung cancer by regulating energy metabolism reprogramming. Cell Death Dis 2023; 14:617. [PMID: 37730658 PMCID: PMC10511739 DOI: 10.1038/s41419-023-06142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Aldehyde dehydrogenase 3A1 (ALDH3A1) is an NAD+-dependent enzyme that is closely related to tumor development. However, its role in non-small-cell lung cancer (NSCLC) has not been elucidated. This study aimed to clarify the mechanism of ALDH3A1 and identify potential therapeutic targets for NSCLC. Here, for the first time, we found that ALDH3A1 expression could be induced by a hypoxic environment in NSCLC. ALDH3A1 was highly expressed in NSCLC tissue, especially in some late-stage patients, and was associated with a poor prognosis. In mechanistic terms, ALDH3A1 enhances glycolysis and suppresses oxidative phosphorylation (OXPHOS) to promote cell proliferation by activating the HIF-1α/LDHA pathway in NSCLC. In addition, the results showed that ALDH3A1 was a target of β-elemene. ALDH3A1 can be downregulated by β-elemene to inhibit glycolysis and enhance OXPHOS, thus suppressing NSCLC proliferation in vitro and in vivo. In conclusion, hypoxia-induced ALDH3A1 is related to the energy metabolic status of tumors and the efficacy of β-elemene, providing a new theoretical basis for better clinical applications in NSCLC.
Collapse
Affiliation(s)
- Yang Chen
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, 110001, Shenyang, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Hongfei Yan
- Department of Medical Oncology, The First Hospital of China Medical University, 110001, Shenyang, China
- Key laboratory of anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, 110001, Shenyang, China
| | - Lirong Yan
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, 110001, Shenyang, China
| | - Ximing Wang
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, 110001, Shenyang, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, 110001, Shenyang, China
- Key laboratory of anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, 110001, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, 110001, Shenyang, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, 110001, Shenyang, China
- Key laboratory of anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, 110001, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, 110001, Shenyang, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, 110001, Shenyang, China
| | - Xuena Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, 110001, Shenyang, China
| | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, 110001, Shenyang, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, 110001, Shenyang, China.
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, 110001, Shenyang, China.
| |
Collapse
|
9
|
Hypoxic Microenvironment-Induced Reduction in PTEN-L Secretion Promotes Non-Small Cell Lung Cancer Metastasis through PI3K/AKT Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6683104. [PMID: 35280516 PMCID: PMC8906955 DOI: 10.1155/2022/6683104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 11/24/2022]
Abstract
Objective Lung cancer is the leading cause of cancer-related deaths worldwide. The aim of this study was to investigate the effects of hypoxic microenvironment on PTEN-L secretion and the effects of PTEN-L on the metastasis of non-small cell lung cancer (NSCLC) and the potential mechanisms. Methods The expression levels of PTEN-L in NSCLC tissues, cells, and cell culture media were detected. The transfection of PTEN-L overexpression construct or HIF-1α-siRNAs was conducted to manipulate the expression of PTEN-L or HIF-1α. NSCLC cells were introduced into 200 μM CoCl2 medium for 72 hours under 37°C to simulate hypoxia. The proliferation and apoptosis of the A549 cells were determined by the Cell Counting Kit-8 assay and Annexin V-FITC/PI-stained flow cytometry assay, respectively. Wound healing assay and transwell invasion assay were used to measure the migration and invasion of A549 cells. The protein expression of PTEN, PTEN-L, PI3K/AKT pathway-related proteins, and HIF-1α was detected by Western blot. Results PTEN and PTEN-L are downregulated in lung cancer tissues and cells. The protein expression of PTEN-L in the culture medium of lung cancer cell lines is decreased. The hypoxic microenvironment inhibits PTEN-L secretion. The low level of PTEN-L promotes cell proliferation, migration, and invasion, as well as inhibits apoptosis of A549 cells. The overexpression of PTEN-L attenuated the activation of the PI3K/AKT pathway by the hypoxic microenvironment. The knockdown of HIF-1α upregulates PTEN-L secretion under hypoxia. Conclusions The hypoxic microenvironment inhibits PTEN-L secretion and thus activates PI3K/AKT pathway to induce proliferation, migration, and invasion promotion, and apoptosis inhibition in NSCLC cells.
Collapse
|
10
|
Liu J, Yang C, Huang XM, Lv PP, Yang YK, Zhao JN, Zhao SY, Sun WJ. Knockdown of FBI-1 Inhibits the Warburg Effect and Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Molecular Targeted Agents via miR-3692/HIF-1α. Front Oncol 2021; 11:796839. [PMID: 34869045 PMCID: PMC8633402 DOI: 10.3389/fonc.2021.796839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 01/20/2023] Open
Abstract
The transcription suppressor factor FBI-1 (the factor that binds to inducer of short transcripts-1) is an important regulator of hepatocellular carcinoma (HCC). In this work, the results showed that FBI-1 promoted the Warburg effect and enhances the resistance of hepatocellular carcinoma cells to molecular targeted agents. Knockdown of FBI-1 via its small-interfering RNA (siRNA) inhibited the ATP level, lactate productions, glucose uptake or lactate dehydrogenase (LDH) activation of HCC cells. Transfection of siFBI-1 also decreased the expression of the Warburg-effect-related factors: hypoxia-inducible factor-1 alpha (HIF-1α), lactate dehydrogenase A (LDHA), or GLUT1, and the epithelial-mesenchymal transition-related factors, Vimentin or N-cadherin. The positive correlation between the expression of FBI-1 with HIF-1α, LDHA, or GLUT1 was confirmed in HCC tissues. Mechanistically, the miR-30c repressed the expression of HIF-1α by binding to the 3'-untranslated region (3'-UTR) of HIF-1α in a sequence-specific manner, and FBI-1 enhanced the expression of HIF-1α and HIF-1α pathway's activation by repressing the expression of miR. By modulating the miR-30c/HIF-1α, FBI-1 promoted the Warburg effect or the epithelial-mesenchymal transition of HCC cells and promoted the resistance of HCC cells to molecular targeted agents.
Collapse
Affiliation(s)
- Juan Liu
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chao Yang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xiao-Mei Huang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Pan-Pan Lv
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ya-Kun Yang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jin-Na Zhao
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Si-Yuan Zhao
- Department of Neurosurgery, Beijing Huicheng Medical Research Institute, Beijing, China
| | - Wan-Jun Sun
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|