1
|
Sheng L, Shen M, Shao D. Total Glycosides of Paeony Activates PI3K/Akt Pathway to Alleviate Cardiomyocyte Hypertrophy Induced by AngII. Cell Biochem Biophys 2025; 83:2059-2066. [PMID: 39821826 DOI: 10.1007/s12013-024-01616-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 01/19/2025]
Abstract
Total glucosides of paeony (TGP) have been investigated for their effects on cardiomyocyte hypertrophy induced by angiotensin II (Ang II). In this study, rat cardiomyocyte H9c2 cells were treated with various doses of TGP (0, 12.5, 25, 50, 100, 200, and 400 μmol/L), and cell viability was assessed using the MTT method to determine an optimal dose. To establish the cardiomyocyte hypertrophy model, Ang II (1 μmol/L) was used. The experimental groups included the control (Ctrl) group, the hypertrophy group (Ang II), the TGP treatment group (TGP+Ang II), and a combined treatment group (TGP+Ang II+LY), where LY294002, a PI3K/Akt inhibitor, was used. The surface area of H9c2 cells was analyzed using image analysis software, and apoptosis was assessed via flow cytometry. Western blotting was employed to evaluate markers related to cell proliferation, cardiac hypertrophy, apoptosis, and autophagy, as well as the phosphorylation of the PI3K/Akt pathway. The results revealed that Ang II inhibited cell viability and increased cell surface area, apoptosis, and autophagy, all of which were significantly reversed by TGP treatment. Moreover, the addition of LY294002 partially attenuated the effects of TGP, reducing cell viability and promoting hypertrophy, apoptosis, and autophagy. Additionally, Ang II reduced PI3K/Akt signaling activity, while TGP restored it. LY treatment reversed the effects of TGP and suppressed the PI3K/Akt pathway. In conclusion, TGP improves cardiomyocyte hypertrophy induced by Ang II by activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Letian Sheng
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Jiangsu, Suzhou, 215000, China
| | - Mengjiao Shen
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Jiangsu, Suzhou, 215000, China
| | - Danyan Shao
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Jiangsu, Suzhou, 215000, China.
| |
Collapse
|
2
|
Liao D, He Y, He B, Zeng S, Cui Y, Li C, Huang H. Inhibiting SNX10 induces autophagy to suppress invasion and EMT and inhibits the PI3K/AKT pathway in cervical cancer. Clin Transl Oncol 2025; 27:2084-2094. [PMID: 39367898 DOI: 10.1007/s12094-024-03715-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/02/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE Cervical cancer (CC) is a prevalent malignancy among women with high morbidity and poor prognosis. Sorting nexin 10 (SNX10) is a newly recognized cancer regulatory factor, while its action on CC progression remains elusive. Hence, this study studied the effect of SNX10 on CC development and investigated the mechanism. METHODS The SNX10 level in CC and the overall survival of CC cases with different SNX10 expressions were determined by bioinformatics analysis in GEPIA. The SNX10 expression in tumor tissues and clinical significance were studied in 64 CC cases. The overall survival was assessed using Kaplan-Meier analysis. The formation of LC3 was evaluated using immunofluorescence. Cell invasion was measured using the Transwell assay. Epithelial-to-mesenchymal transition (EMT) was determined by observing cell morphology and assessing EMT marker levels. A xenograft tumor was constructed to evaluate tumor growth. RESULTS SNX10 was elevated in CC tissues and cells, and the CC cases with high SNX10 levels exhibited poor overall survival. Besides, SNX10 correlated with the FIGO stage, lymph node invasion, and stromal invasion of CC. SNX10 silencing induced CC cell autophagy and suppressed CC cell invasion and EMT. Meanwhile, silenced SNX10 could suppress invasion and EMT via inducing autophagy. Furthermore, SNX10 inhibition suppressed the PI3K/AKT pathway. Moreover, silenced SNX10 restrained the tumor growth, autophagy, and EMT of CC in vivo. CONCLUSION SNX10 was enhanced in CC and correlated with poor prognosis. Silenced SNX10 induced autophagy to suppress invasion and EMT and inhibited the PI3K/AKT pathway in CC, making SNX10 a valuable molecule for CC therapy.
Collapse
Affiliation(s)
- Dan Liao
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| | - Yanxian He
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Bin He
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Saitian Zeng
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Yejia Cui
- Department of Clinical Laboratory, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Cuifen Li
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Haohai Huang
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China.
- Department of Clinical Pharmacy, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| |
Collapse
|
3
|
Zheng Y, Feng N, Li C, Li Z. Natural products target programmed cell death signaling mechanisms to treat colorectal cancer. Front Pharmacol 2025; 16:1565332. [PMID: 40342991 PMCID: PMC12058791 DOI: 10.3389/fphar.2025.1565332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
As a highly prevalent gastrointestinal malignant tumor, colorectal cancer poses a serious challenge in terms of increasing morbidity and mortality and late diagnosis due to the invisibility of the disease. Although existing therapies are diverse but limited in efficacy, the mechanism of programmed cell death (PCD) has become a focus of research due to its central role in maintaining body homeostasis and regulating tumor progression. Multimodal cell death pathways, such as apoptosis, autophagy, pyroptosis and ferroptosis, have shown unique advantages in inhibiting the proliferation and migration of colorectal cancer cells and enhancing the sensitivity to chemotherapy by responding to internal and external environmental stimuli. In recent years, natural products have risen to prominence by virtue of their multi-target synergistic effects and chemo-sensitizing properties, and have opened up a new direction for colorectal cancer treatment by precisely regulating the PCD pathway. In this paper, we searched PubMed, Web of Science and CNKI databases for relevant studies in the last 10 years using the keywords (Colorectal cancer) and (programmed cell death) and natural products. This work retrieved 59 studies (55 from the past 5 years and 4 from the past 10 years) to reveal the mechanism of action of natural products targeting PCD, aiming to provide theoretical support and practical guidance for the optimization of clinical therapeutic strategies and the development of innovative drugs.
Collapse
Affiliation(s)
- Ya Zheng
- The Second Gastroenterology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Na Feng
- Department of Rehabilitation Medicine, Linyi Maternal and Child Health Center Hospital, Linyi, Shandong, China
| | - Canglin Li
- Medical Management Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zuoqiang Li
- Department of Traditional Chinese Medicine, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| |
Collapse
|
4
|
Simoni MK, Negatu SG, Park JY, Mani S, Arreguin MC, Amses KR, Huh DD, Mainigi M, Jurado KA. Type I interferon exposure of an implantation-on-a-chip device alters invasive extravillous trophoblast function. Cell Rep Med 2025; 6:101991. [PMID: 40054459 PMCID: PMC11970386 DOI: 10.1016/j.xcrm.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/12/2025]
Abstract
Inappropriate type I interferon (IFN) signaling during embryo implantation and placentation is linked to poor pregnancy outcomes. Here, we evaluate the consequence of elevated type I IFN exposure on implantation using a human implantation in an organ-on-a-chip device. We reveal that type I IFN reduces extravillous trophoblast (EVT) invasion capacity. Analyzing single-cell transcriptomes, we uncover that IFN truncates invasive EVT emergence in the implantation-on-a-chip device by stunting EVT epithelial-to-mesenchymal transition. Disruptions to the epithelial-to-mesenchymal transition are associated with the pathogenesis of preeclampsia, a life-threatening disorder of pregnancy. Strikingly, IFN stimulation induces genes associated with increased preeclampsia risk in EVTs. These dysregulated EVT phenotypes ultimately reduce EVT-mediated endothelial cell vascular remodeling in the implantation-on-a-chip device. Overall, our work implicates unwarranted type I IFN as a maternal disturbance that can result in abnormal EVT function that could trigger preeclampsia.
Collapse
Affiliation(s)
- Michael K Simoni
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seble G Negatu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sneha Mani
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Montserrat C Arreguin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Kellie A Jurado
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Alexander AK, Rodriguez KF, Chen YY, Amato C, Estermann MA, Nicol B, Xu X, Yao HHC. Single-nucleus multiomics reveals the gene regulatory networks underlying sex determination of murine primordial germ cells. eLife 2025; 13:RP96591. [PMID: 40063068 PMCID: PMC11893106 DOI: 10.7554/elife.96591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
Accurate specification of female and male germ cells during embryonic development is critical for sexual reproduction. Primordial germ cells (PGCs) are the bipotential precursors of mature gametes that commit to an oogenic or spermatogenic fate in response to sex-determining cues from the fetal gonad. The critical processes required for PGCs to integrate and respond to signals from the somatic environment in gonads are not well understood. In this study, we developed the first single-nucleus multiomics map of chromatin accessibility and gene expression during murine PGC development in both XX and XY embryos. Profiling of cell-type-specific transcriptomes and regions of open chromatin from the same cell captured the molecular signatures and gene networks underlying PGC sex determination. Joint RNA and ATAC data for single PGCs resolved previously unreported PGC subpopulations and cataloged a multimodal reference atlas of differentiating PGC clusters. We discovered that regulatory element accessibility precedes gene expression during PGC development, suggesting that changes in chromatin accessibility may prime PGC lineage commitment prior to differentiation. Similarly, we found that sexual dimorphism in chromatin accessibility and gene expression increased temporally in PGCs. Combining single-nucleus sequencing data, we computationally mapped the cohort of transcription factors that regulate the expression of sexually dimorphic genes in PGCs. For example, the gene regulatory networks of XX PGCs are enriched for the transcription factors, TFAP2c, TCFL5, GATA2, MGA, NR6A1, TBX4, and ZFX. Sex-specific enrichment of the forkhead-box and POU6 families of transcription factors was also observed in XY PGCs. Finally, we determined the temporal expression patterns of WNT, BMP, and RA signaling during PGC sex determination, and our discovery analyses identified potentially new cell communication pathways between supporting cells and PGCs. Our results illustrate the diversity of factors involved in programming PGCs toward a sex-specific fate.
Collapse
Affiliation(s)
- Adriana K Alexander
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Karina F Rodriguez
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Yu-Ying Chen
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Ciro Amato
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Martin A Estermann
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Barbara Nicol
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Xin Xu
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| | - Humphrey HC Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle ParkDurhamUnited States
| |
Collapse
|
6
|
Rakoczy K, Szymańska N, Stecko J, Kisiel M, Sleziak J, Gajewska-Naryniecka A, Kulbacka J. The Role of RAC2 and PTTG1 in Cancer Biology. Cells 2025; 14:330. [PMID: 40072059 PMCID: PMC11899714 DOI: 10.3390/cells14050330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Several molecular pathways are likely involved in the regulation of cancer stem cells (CSCs) via Ras-associated C3 botulinum toxin substrate 2, RAC2, and pituitary tumor-transforming gene 1 product, PTTG1, given their roles in cellular signaling, survival, proliferation, and metastasis. RAC2 is a member of the Rho GTPase family and plays a crucial role in actin cytoskeleton dynamics, reactive oxygen species production, and cell migration, contributing to epithelial-mesenchymal transition (EMT), immune evasion, and therapy resistance. PTTG1, also known as human securin, regulates key processes such as cell cycle progression, apoptosis suppression, and EMT, promoting metastasis and enhancing cancer cell survival. This article aims to describe the molecular pathways involved in the proliferation, invasiveness, and drug response of cancer cells through RAC2 and PTTG1, aiming to clarify their respective roles in neoplastic process dependencies. Both proteins are involved in critical signaling pathways, including PI3K/AKT, TGF-β, and NF-κB, which facilitate tumor progression by modulating CSC properties, angiogenesis, and immune response. This review highlights the molecular mechanisms by which RAC2 and PTTG1 influence tumorigenesis and describes their potential and efficacy as prognostic biomarkers and therapeutic targets in managing various neoplasms.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (N.S.); (J.S.); (M.K.); (J.S.)
| | - Natalia Szymańska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (N.S.); (J.S.); (M.K.); (J.S.)
| | - Jakub Stecko
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (N.S.); (J.S.); (M.K.); (J.S.)
| | - Michał Kisiel
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (N.S.); (J.S.); (M.K.); (J.S.)
| | - Jakub Sleziak
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (N.S.); (J.S.); (M.K.); (J.S.)
| | - Agnieszka Gajewska-Naryniecka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406 Vilnius, Lithuania
| |
Collapse
|
7
|
Duan S, Tian Z, Hu R, Long H. NEDD4L inhibits epithelial-mesenchymal transition in gastric cancer by mediating BICC1 ubiquitination. Kaohsiung J Med Sci 2025; 41:e12924. [PMID: 39717922 PMCID: PMC11827545 DOI: 10.1002/kjm2.12924] [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: 09/13/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a critical stage in the metastasis of gastric cancer (GC). Further clarification of the EMT process in GC is still needed. This study examined the effects of the NEDD4L/BICC1 axis on GC proliferation and the EMT process. Thirty GC patients were enrolled in this study to assess the expression of BICC1 and NEDD4L in tumor samples. A xenograft tumor model in mice was created to investigate BICC1's function in vivo. The proliferation, migration, and invasion of GC cells were evaluated using colony formation, transwell, and wound healing assays. Western blot determined the expression levels of EMT-associated proteins. Co-immunoprecipitation (Co-IP) elucidated the mechanism by which NEDD4L regulates BICC1. BICC1 was found to be overexpressed in tumors. Additionally, BICC1 knockdown inhibited the growth of GC cells in vivo and prevented their migration, invasion, proliferation, and EMT. Furthermore, BICC1 activated the PI3K/AKT pathway, which facilitated cancer progression. Tumor tissues and GC cells exhibited low expression levels of NEDD4L. Conversely, NEDD4L overexpression promoted the ubiquitination and degradation of BICC1 protein, thereby inhibiting GC cell proliferation, migration, invasion, and EMT processes. Our study demonstrated that NEDD4L acts as a tumor suppressor in GC, while BICC1 functions as a pro-tumorigenic factor. The NEDD4L/BICC1 axis plays a significant role in the metastasis and progression of GC.
Collapse
Affiliation(s)
- Shaoyi Duan
- Hunan University of MedicineHuaihuaHunan ProvincePeople's Republic of China
| | - Zhiliang Tian
- Hunan University of MedicineHuaihuaHunan ProvincePeople's Republic of China
| | - Rong Hu
- Hunan University of MedicineHuaihuaHunan ProvincePeople's Republic of China
| | - Heng Long
- Hunan University of MedicineHuaihuaHunan ProvincePeople's Republic of China
| |
Collapse
|
8
|
Wang C, Guo J, Zhang Y, Zhou S, Jiang B. Cuproptosis-Related Gene FDX1 Suppresses the Growth and Progression of Colorectal Cancer by Retarding EMT Progress. Biochem Genet 2025; 63:775-788. [PMID: 38520567 PMCID: PMC11832605 DOI: 10.1007/s10528-024-10784-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/28/2024] [Indexed: 03/25/2024]
Abstract
Colorectal cancer (CRC) is a usual cancer and a kind of lethiferous cancer. Cuproptosis-related gene ferredoxin 1 (FDX1) has been discovered to act as a suppressor, thereby suppressing some cancers' progression. But, the regulatory functions of FDX1 in CRC progression keep vague. In this work, at first, through TCGA database, it was revealed that FDX1 exhibited lower expression in COAD (colon adenocarcinoma) tissues, and CRC patients with lower FDX1 expression had worse prognosis. Furthermore, FDX1 expression was verified to be down-regulated in CRC tissues (n = 30) and cells. It was further uncovered that FDX1 expression was positively correlated with CDH1 and TJP1 (epithelial marker), and negatively correlated with CDH2, TWIST1, and FN1 (stromal marker), suggesting that FDX1 was closely associated with the epithelial-mesenchymal transition (EMT) progress. Next, it was demonstrated that overexpression of FDX1 suppressed cell viability, invasion, and migration in CRC. Furthermore, it was verified that FDX1 retarded the EMT progress in CRC. Lastly, through rescue assays, the inhibited CRC progression mediated by FDX1 overexpression was rescued by EGF (EMT inducer) treatment. At last, it was uncovered that the tumor growth and metastasis were relieved after FDX1 overexpression, but these changes were reversed after EGF treatment. In conclusion, FDX1 inhibited the growth and progression of CRC by inhibiting EMT progress. This discovery hinted that FDX1 may act as an effective candidate for CRC treatment.
Collapse
Affiliation(s)
- Chao Wang
- Department of Internal Medicine Oncology, Chaohu Hospital of Anhui Medical University, No. 64, Chaohu North Road, Juchao District, Chaohu, 238000, Anhui, China.
| | - Jingjing Guo
- Department of Internal Medicine Oncology, Chaohu Hospital of Anhui Medical University, No. 64, Chaohu North Road, Juchao District, Chaohu, 238000, Anhui, China
| | - Yun Zhang
- Department of Internal Medicine Oncology, Chaohu Hospital of Anhui Medical University, No. 64, Chaohu North Road, Juchao District, Chaohu, 238000, Anhui, China
| | - Shusheng Zhou
- Department of Internal Medicine Oncology, Chaohu Hospital of Anhui Medical University, No. 64, Chaohu North Road, Juchao District, Chaohu, 238000, Anhui, China
| | - Bing Jiang
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China
| |
Collapse
|
9
|
Han M, Zhou X, Cheng H, Qiu M, Qiao M, Geng X. Chitosan and hyaluronic acid in colorectal cancer therapy: A review on EMT regulation, metastasis, and overcoming drug resistance. Int J Biol Macromol 2025; 289:138800. [PMID: 39694373 DOI: 10.1016/j.ijbiomac.2024.138800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Up to 90% of cancer-related fatalities could be attributed to metastasis. Therefore, understanding the mechanisms that facilitate tumor cell metastasis is beneficial for improving patient survival and results. EMT is considered the main process involved in the invasion and spread of CRC. Essential molecular components like Wnt, TGF-β, and PI3K/Akt play a role in controlling EMT in CRC, frequently triggered by various factors such as Snail, Twist, and ZEB1. These factors affect not only the spread of CRC but also determine the reaction to chemotherapy. The influence of non-coding RNAs, especially miRNAs and lncRNAs, on the regulation of EMT is clear in CRC. Exosomes, involved in cell-to-cell communication, can affect the TME and metastasis of CRC. Pharmacological substances and nanoparticles demonstrate promise as efficient modulators of EMT in CRC. Chitosan and HA are two major carbohydrate polymers with considerable potential in inhibiting CRC. Chitosan and HA can be employed to modify nanoparticles to enhance cargo transport for reducing CRC. Additionally, chitosan and HA-modified nanocarriers, which can be utilized as potential approaches in suppressing EMT and reversing drug resistance in CRC, can inhibit EMT and chemoresistance, crucial components in tumorigenesis.
Collapse
Affiliation(s)
- Mingming Han
- Department of Pharmacy and Medical Devices, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Xi Zhou
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Hang Cheng
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Mengru Qiu
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Meng Qiao
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Xiao Geng
- Department of Party Committee Office, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| |
Collapse
|
10
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [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: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
11
|
Li Z, Li J, Cao Q, Shen T, Wang Y, He H, Tong M. Transcription factor TCF7L1 targeting HSPB6 is involved in EMT and PI3K/AKT/mTOR pathways in bladder cancer. J Biol Chem 2025; 301:108024. [PMID: 39608715 PMCID: PMC11728895 DOI: 10.1016/j.jbc.2024.108024] [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: 03/26/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024] Open
Abstract
Bladder cancer is notorious for its high recurrence and costly treatment burden, prompting a search for novel therapeutic targets. Our study focuses on HSPB6, a small heat shock protein whose reduced expression in bladder cancer suggests a role in tumor biology. Using an integrative approach of bioinformatics, RNA sequencing, and cell-based assays, we show that HSPB6 upregulation inhibits cancer cell proliferation and metastasis while promoting apoptosis. Moreover, TCF7L1-mediated upregulation of HSPB6 leads to suppression of the PI3K/AKT/mTOR signaling pathway, a key driver of cancer progression. These results position HSPB6 as a compelling target for bladder cancer therapy, and its regulatory role in the PI3K/AKT/mTOR axis underscores its therapeutic potential. Our findings pave the way for future investigations into HSPB6-centered treatment strategies.
Collapse
Affiliation(s)
- Zizhi Li
- Department of Medicine, Soochow University, Soochow, Jiangsu, China; Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Junyi Li
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Qingfei Cao
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Tong Shen
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yingjie Wang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Haoyang He
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ming Tong
- Department of Medicine, Soochow University, Soochow, Jiangsu, China; Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
12
|
Qin H, Wang Q, Xu J, Zeng H, Liu J, Yu F, Yang J. Integrative analysis of anoikis-related genes prognostic signature with immunotherapy and identification of CDKN3 as a key oncogene in lung adenocarcinoma. Int Immunopharmacol 2024; 143:113282. [PMID: 39383787 DOI: 10.1016/j.intimp.2024.113282] [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: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
Anoikis, a form of programmed cell death induced by loss of cell contact, is closely associated with tumor invasion and metastasis, making it highly significant in lung cancer research. We examined the expression patterns and prognostic relevance of Anoikis-related genes (ARGs) in lung adenocarcinoma (LUAD) using the TCGA-LUAD database. This study identified molecular subtypes associated with Anoikis in LUAD and conducted functional enrichment analyses. We constructed an ARG risk score using univariate least absolute shrinkage and selection operator (LASSO) Cox regression, validated externally with GEO datasets and clinical samples. The clinical applicability of the prognostic model was evaluated using nomograms, calibration curves, decision curve analysis (DCA), and time-dependent AUC assessments. We identified four prognostically significant genes (PLK1, SLC2A1, CDKN3, PHLDA2) and two ARG-related molecular subtypes. ARGs were generally upregulated in LUAD and correlated with multiple pathways including the cell cycle and DNA replication. The prognostic model indicated that the low-risk group had better outcomes and significant correlations with clinicopathological features, tumor microenvironment, immune therapy responses, drug sensitivity, and pan-RNA epigenetic modification-related genes. Patients with low-risk LUAD were potential beneficiaries of immune checkpoint inhibitor (ICI) therapy. Prognostic ARGs' distribution and expression across various immune cell types were further analyzed using single-cell RNA sequencing. The pivotal role of CDKN3 in LUAD was confirmed through qRT-PCR and gene knockout experiments, demonstrating that CDKN3 knockdown inhibits tumor cell proliferation, migration, and invasion. Additionally, we constructed a ceRNA network involving CDKN3/hsa-miR-26a-5p/SNHG6, LINC00665, DUXAP8, and SLC2A1/hsa-miR-218-5p/RNASEH1-AS1, providing new insights for personalized and immune therapy decisions in LUAD patients.
Collapse
Affiliation(s)
- Haotian Qin
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Qichang Wang
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Juan Xu
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei 238001, China
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Fei Yu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China.
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| |
Collapse
|
13
|
Rajendran D, Oon CE. Navigating therapeutic prospects by modulating autophagy in colorectal cancer. Life Sci 2024; 358:123121. [PMID: 39389340 DOI: 10.1016/j.lfs.2024.123121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Colorectal cancer (CRC) remains a leading cause of death globally despite the improvements in cancer treatment. Autophagy is an evolutionarily conserved lysosomal-dependent degradation pathway that is critical in maintaining cellular homeostasis. However, in cancer, autophagy may have conflicting functions in preventing early tumour formation versus the maintenance of advanced-stage tumours. Defective autophagy has a broad and dynamic effect not just on cancer cells, but also on the tumour microenvironment which influences tumour progression and response to treatment. To add to the layer of complexity, somatic mutations in CRC including tumour protein p53 (TP53), v-raf murine sarcoma viral oncogene homolog B1 (BRAF), Kirsten rat sarcoma viral oncogene homolog (KRAS), and phosphatase and tensin homolog (PTEN) can render chemoresistance by promoting a pro-survival advantage through autophagy. Recent studies have also reported autophagy-related cell deaths that are distinct from classical autophagy by employing parts of the autophagic machinery, which impacts strategies for autophagy regulation in cancer therapy. This review discusses the molecular processes of autophagy in the evolution of CRC and its role in the tumour microenvironment, as well as prospective therapeutic methods based on autophagy suppression or promotion. It also highlights clinical trials using autophagy modulators for treating CRC, underscoring the importance of autophagy regulation in CRC therapy.
Collapse
Affiliation(s)
- Deepa Rajendran
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| |
Collapse
|
14
|
Xu J, Zhang Y, Chen H, Zhang J, Zhu J, He Y, Cui G. Up-regulation of LPCAT1 is correlated with poor prognosis and promotes tumor progression in glioblastoma. Cytotechnology 2024; 76:735-748. [PMID: 39435418 PMCID: PMC11490591 DOI: 10.1007/s10616-024-00650-1] [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: 06/20/2023] [Accepted: 07/29/2024] [Indexed: 10/23/2024] Open
Abstract
Glioblastoma (GBM) is a cancer with high malignancy because of its rapid proliferation and high metastatic ability. LPCAT1 is reported to play a tumor-promoting role in multiple cancers, but its precise molecular mechanism in GBM remains to be further explored. We aim to explore the biological role of LPCAT1 in GBM. In this study, the expression of LPCAT1 and its correlation with clinicopathological characteristics of GBM patients were analyzed based on The Cancer Genome Atlas (TCGA) dataset. Kaplan-Meier approach was adopted for plotting survival curves for patients showing different expression levels of LPCAT1. Meanwhile, LPCAT1 expression within 50 GBM tumor tissues and 30 non-tumor clinical samples was analyzed by qRT-PCR and western blot assays, respectively. Later, LPCAT1's effect on GBM tumorigenesis was analyzed in vivo and in vitro by CCK8, EdU proliferation, clone forming, scratch, TUNEL assays, and subcutaneous xenograft experiments. As a result, LPCAT1 expression elevated within GBM tumor tissues and cells. Overexpression of LPCAT1 enhanced GBM cell growth, invasion and migration, while accelerating cell cycle progression. LPCAT1 silencing significantly inhibited cell motility and proliferation in vivo and in vitro, and arrested U251 cells at G0/G1 phase. Moreover, LPCAT1 might play a role in GBM progression by activating the p-AKT-MYC signaling pathway. LPCAT1 activated AKT, which were synchronously up-regulated MYC to accelerate cancer progression. Knockdown of LPCAT1 induced the opposite changes to repress the viability and motility of GBM cells. LPCAT1 contributed to the progression of GBM by participating in the p-AKT-MYC axis.
Collapse
Affiliation(s)
- Jin Xu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006 China
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, 223800 China
| | - Yuan Zhang
- Department of Radiology, Suzhou Guangji Hospital, Suzhou, 215100 China
| | - Honglin Chen
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, 223800 China
| | - Jianyong Zhang
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, 223800 China
| | - Jie Zhu
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, 223800 China
| | - Yuchao He
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, 223800 China
| | - Gang Cui
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006 China
| |
Collapse
|
15
|
Shen C, Huang P, Xie W, Ni X, Gao J. Sini decoction-polysaccharide compound regulates proliferation, apoptosis, and glycolysis of liver cancer cells through PHLDA2/ANXA2. Transl Cancer Res 2024; 13:5574-5587. [PMID: 39525010 PMCID: PMC11543045 DOI: 10.21037/tcr-24-1625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Background Sini decoction (SND), a popular formula from traditional Chinese medicine (TCM), plays a critical role in the treatment of liver disease. Its protective effect for the heart against cardiovascular diseases is well documented. However, its effects and pharmacological mechanisms for the liver remain unclear. This study aimed to clarify the effect and mechanism of the SND-polysaccharide compound (SNDPC) on hepatocellular carcinoma (HCC). Methods Different genes affected by SNDPC in HCC were analyzed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Databases including Multi-Experiment Matrix (MEM), HCCDB, LinkedOmics, and Gene Expression Profiling Interactive Analysis (GEPIA) were used to determine the correlation between PHLDA2 and ANXA2. Cell proliferation and viability were identified using Cell Counting Kit-8 (CCK-8). Cell apoptosis was estimated using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay and Western blotting. Glycolysis was determined by measuring glucose uptake, lactate concentration, extracellular acidification rate (ECAR), and the expressions of LHDA, HK2, and PKM2. The binding between PHLDA2 and ANXA2 was identified by coimmunoprecipitation. Results SNDPC significantly weakened cell proliferation, facilitated cell apoptosis, and suppressed glycolysis by reducing glucose uptake, lactate concentration, ECAR, and the expressions of LDHA, HK2, and PKM2 in HCC cells. Furthermore, PHLDA2 was predicted to bind to ANXA2, which was confirmed by coimmunoprecipitation. SNDPC reduced the expressions of PHLDA2 and ANXA2 in HCCLM3 cells, and PHLDA2 silencing decreased the proliferation of cells, promoted cell apoptosis, and inhibited glycolysis of HCCLM3 cells while reversing the overexpression of PHLDA2. Conclusions SNDPC suppressed proliferation and glycolysis while accelerating the apoptosis of HCC cells through PHLDA2/ANXA2.
Collapse
Affiliation(s)
- Churan Shen
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Peipei Huang
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Wuji Xie
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xing Ni
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Jingdong Gao
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| |
Collapse
|
16
|
Xiang Z, Yu S, Xu Y, Xiong H, Hu D, Li Q, Wu Z. CH25H Promotes Autophagy and Regulates the Malignant Progression of Laryngeal Squamous Cell Carcinoma Through the PI3K-AKT Pathway. Cancer Med 2024; 13:e70312. [PMID: 39428922 PMCID: PMC11491687 DOI: 10.1002/cam4.70312] [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: 01/09/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Laryngeal squamous cell carcinoma (LSCC) is a type of cancer of the respiratory tract that often presents with subtle symptoms at the early stage and is susceptible to recurrence and metastasis. MATERIALS AND METHODS To find out key regulatory genes involved in LSCC development, we downloaded LSCC-related sequencing datasets for bioinformatics analysis. WGCNA was performed on GSE142083 and differential analysis was conducted on GSE51985 and TCGA-HNSC. Intersection genes were taken from the above three datasets. To confirm the function of genes, we overexpressed and knocked down genes in cells and treated them with autophagy agonist Rapamycin and PI3K-AKT pathway inhibitor. At the cellular level, the expression of CH25H, autophagy-related proteins (LC3 I, LC3 II, p62, and Beclin 1), and PI3K-AKT pathway-related proteins (PI3K, AKT, and p-AKT) were assessed via Western blot; the mRNA level of CH25H was evaluated through qRT-PCR; the cell activity was examined by CCK8; the apoptosis was assessed through flow cytometry; and the cell migration and invasion were assessed through wound healing and Transwell assays. RESULTS Through bioinformatics analysis, we screened 7 genes (CH25H, NELL2, STC2, TMEM158, ZIC2, HOXD11, and HOXD10). Ultimately, CH25H was selected for follow-up experiments. By detecting CH25H expression in human immortalized keratinocytes (HaCaT) and LSCC cells (Tu-686, SNU899, and AMC-HN-8), it was found out that CH25H expression was higher in HaCaT cells than in LSCC cells. To elucidate the role of CH25H in LSCC development, we overexpressed CH25H in Tu-686 cells and downregulated its expression in AMC-HN-8 cells. CH25H was revealed to reduce the proliferation, activity, invasion, and migration of LSCC cells while increasing their apoptosis levels. Significant changes were also observed in the expressions of autophagy- and PI3K-AKT pathway-related proteins. To further investigate the roles of autophagy and the PI3K-AKT pathway in LSCC development, we respectively employed autophagy agonists and inhibitors targeting the PI3K-AKT pathway to intervene the cells, and found that CH25H regulated the PI3K-AKT pathway to promote autophagy, thus enhancing the apoptosis of LSCC cells. We further investigated CH25H's impact on tumor growth, autophagy, and the PI3K-AKT pathway at the animal level and found that CH25H promoted autophagy of LSCC cells and inhibited the PI3K-AKT pathway, and ultimately inhibiting the progression of LSCC. CONCLUSIONS In summary, CH25H promotes autophagy and affects the malignant progression of LSCC through the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Zhenfei Xiang
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Senquan Yu
- Department of OncologyThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yuxin Xu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Huacai Xiong
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Danfei Hu
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Qun Li
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Zhenhua Wu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
17
|
Sha Y, Zhuang H, Shi J, Ge S, He S, Wang Y, Ma L, Guo H, Cheng H. B3GALT4 modulates tumor progression and autophagy by AKT/mTOR signaling pathway in breast cancer. Discov Oncol 2024; 15:488. [PMID: 39331217 PMCID: PMC11436681 DOI: 10.1007/s12672-024-01371-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND β-1,3-Galactosyltransferase-4 (B3GALT4), a member of the β-1,3-galactosyltransferase gene family, is essential to the development of many malignancies. However, its biological function in breast cancer is still unknown. METHOD Publically accessible datasets, as well as quantitative real-time PCR, western blot, and immunohistochemistry on our patient cohort were used to investigate the expression levels of B3GALT4 in breast cancer. The correlation of B3GALT4 expression with clinical histopathological data and mortality in breast cancer patients was investigated. The effects of B3GALT4 in breast cancer in vitro and in vivo were investigated. RNA-seq, western blot, autophagolysosomes, and the fluorescence intensity of LC3 were used to explore the effects of B3GALT4 on autophagy. Western blot and gene set enrichment analysis (GSEA) were used to identify the AKT/mTOR pathway. RESULTS B3GALT4 was significantly overexpressed in breast cancer tissues and was positively correlated with some aspects of clinicopathological status and poor prognosis. B3GALT4 overexpression significantly promoted cell proliferation, migration, and invasion, both in vitro and in vivo. B3GALT4 inhibition suppressed breast cancer cell proliferation, migration, and invasion in vitro. Suppression of B3GALT4 triggered autophagy and hindered the AKT/mTOR signaling pathway. CONCLUSION According to the present research, B3GALT4 blocked autophagy via the AKT/mTOR pathway and accelerated the growth of breast cancer. B3GALT4 may be an effective target for patients with breast cancer.
Collapse
Affiliation(s)
- Yongliang Sha
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Huijie Zhuang
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Jin Shi
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Song Ge
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Shiqing He
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Yiqiu Wang
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Li Ma
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Hao Guo
- Department of General Surgery, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China.
| | - Hui Cheng
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China.
| |
Collapse
|
18
|
Wei M, Yang X, Yang X, Huang Y, Yuan Z, Huang J, Wei J, Tian L. MLPH regulates EMT in pancreatic adenocarcinoma through the PI3K-AKT signaling pathway. J Cancer 2024; 15:5828-5838. [PMID: 39308678 PMCID: PMC11414609 DOI: 10.7150/jca.94573] [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: 01/22/2024] [Accepted: 08/02/2024] [Indexed: 09/25/2024] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is an extremely malignant tumor, and most patients develop postoperative metastases. Melanophilin (MLPH) is involved in the progression of various tumors, but its molecular mechanisms and role in pancreatic cancer progression are unknown. In this study, differential MLPH expression in cancer tissues and the adjacent tissues was evaluated using the Gene Expression Profiling Interaction Analysis 2 (GEPIA 2) and Human Protein Atlas (HPA) databases. The role of MLPH in PAAD proliferation, invasion, and migration in vitro was explored via clone formation, Cell Counting Kit-8 assay, Transwell assay, and western blot. The in vivo validation of function was performed using a metastatic nude mouse model. The result showed that the pancreatic cancer tissues had significantly higher MLPH expression levels than the noncancerous pancreatic tissues. MLPH expression changes were related to PAAD cell proliferation, invasion, and migration. The western blotting demonstrated that PAAD cells had reduced Epithelial-mesenchymal transition (EMT)-related marker expression. Furthermore, overexpressing MLPH enhanced cell proliferation, migration, and invasion, and increased EMT-related marker expression. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that the molecular mechanism underlying the effect of MLPH on PAAD was significantly related to the PI3K-AKT pathway. LY294002 blocked the MLPH overexpression-mediated enhanced cell invasion and migration and inhibited EMT-associated marker expression. Conversely, 740Y-P reversed the inhibitory effects of MLPH downregulation and led to cell migration, invasion, and EMT. MLPH regulated EMT to mediate PAAD cell invasive migration through the PI3K-AKT pathway. The results indicated that MLPH is a possible target for blocking PAAD metastasis.
Collapse
Affiliation(s)
- Mengda Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Xi Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Xiaoying Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Yanqing Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Zhenmin Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Junjie Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Junren Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Lei Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| |
Collapse
|
19
|
Wang C, Zhao Y, Zhang S, Du M, He G, Tan S, Li H, Zhang D, Cheng L. Single-cell RNA sequencing reveals the heterogeneity of MYH11+ tumour-associated fibroblasts between left-sided and right-sided colorectal cancer. J Cell Mol Med 2024; 28:e70102. [PMID: 39294858 PMCID: PMC11410558 DOI: 10.1111/jcmm.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
Colorectal cancer (CRC) exhibits considerable heterogeneity on tumour location. However, there is still a lack of comprehensive annotation regarding the characteristics and differences between the left-sided (L-CRC) and right-sided (R-CRC) CRC. Here, we performed single-cell RNA sequencing (scRNA-seq) on immune and stromal cells from 12 L-CRC and 10 R-CRC patients. We found that L-CRC exhibited stronger tumour invasion and poor prognosis compared with R-CRC. In addition, functional enrichment analysis of a normal cohort showed that fibroblasts of left colon are associated with tumour-related pathways. This suggested that the heterogeneity observed in both L-CRC and R-CRC may be influenced by the specific location within the colon itself. Further, we identified a potentially novel MYH11+ cancer-associated fibroblast (CAF) subset predominantly enriched in L-CRC. Moreover, we found that MYH11+ CAFs may promote tumour migration via interacting with macrophages, and was associated with poor prognosis in CRC. In summary, our study revealed the crucial role of MYH11+ CAFs in predicting a poor prognosis, thereby contributing valuable insights to the exploration of heterogeneity in L-CRC and R-CRC.
Collapse
Affiliation(s)
- Chao Wang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Yue Zhao
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Sainan Zhang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Meiyu Du
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Guanzhi He
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Senwei Tan
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Hailong Li
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Duoyi Zhang
- The 2nd Affiliated Hospital of Harbin Medical University, Harbin Medical UniversityHarbinHeilongjiangChina
| | - Liang Cheng
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHeilongjiangChina
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and TherapyHarbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
20
|
Yan Y, Yu W, Guo M, Zhu N, Chen X, Li N, Zhong C, Wang G. Autophagy regulates apoptosis of colorectal cancer cells based on signaling pathways. Discov Oncol 2024; 15:367. [PMID: 39182013 PMCID: PMC11344751 DOI: 10.1007/s12672-024-01250-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer is a common malignant tumor of the digestive system. Its morbidity and mortality rank among the highest in the world. Cancer development is associated with aberrant signaling pathways. Autophagy is a process of cell self-digestion that maintains the intracellular environment and has a bidirectional regulatory role in cancer. Apoptosis is one of the important death programs in cancer cells and is able to inhibit cancer development. Studies have shown that a variety of substances can regulate autophagy and apoptosis in colorectal cancer cells through signaling pathways, and participate in the regulation of autophagy on apoptosis. In this paper, we focus on the relevant research on autophagy in colorectal cancer cells based on the involvement of related signaling pathways in the regulation of apoptosis in order to provide new research ideas and therapeutic directions for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yuwei Yan
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Wenyan Yu
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Min Guo
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Naicheng Zhu
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Xiudan Chen
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Nanxin Li
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Chen Zhong
- Research Center for Differentiation and Development of CM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenisis, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Guojuan Wang
- Oncology Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, No.445, Bayi Avenue, Nanchang, 330006, China.
| |
Collapse
|
21
|
Zhao J, Pan X, Wang Z, Chen Y, Liu D, Shen Y, Wei X, Xu C, Zhang X, Hu X, Chen J, Zhao J, Tang B, Sun G, Shen P, Liu Z, Zeng H, Liang J. Epigenetic modification of PHLDA2 is associated with tumor microenvironment and unfavorable outcome of immune checkpoint inhibitor-based therapies in clear cell renal cell carcinoma. Eur J Med Res 2024; 29:378. [PMID: 39033192 PMCID: PMC11264912 DOI: 10.1186/s40001-024-01939-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND A substantial proportion of patients with metastatic clear cell renal cell carcinoma (ccRCC) cannot derive benefit from immune checkpoint inhibitor (ICI) plus anti-angiogenic agent combination therapy, making identification of predictive biomarkers an urgent need. The members of pleckstrin homology-like domain family A (PHLDA) play critical roles in multiple cancers, whereas their roles in ccRCC remain unknown. METHODS Transcriptomic, clinical, genetic alteration and DNA methylation data were obtained for integrated analyses from TCGA database. RNA sequencing was performed on 117 primary tumors and 79 normal kidney tissues from our center. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis, gene set enrichment analysis were performed to explore transcriptomic features. Data from three randomized controlled trials (RCT), including CheckMate025, IMmotion151, JAVELIN101, were obtained for validation. RESULTS Members of PHLDA family were dysregulated in pan-cancer. Elevated PHLDA2 expression was associated with adverse clinicopathologic parameters and worse prognosis in ccRCC. Aberrant DNA hypomethylation contributed to up-regulation of PHLDA2. An immunosuppressive microenvironment featured by high infiltrates of Tregs and cancer-associated fibroblasts, was observed in ccRCC with higher PHLDA2 expression. Utilizing data from three RCTs, the association of elevated PHLDA2 expression with poor therapeutic efficacy of ICI plus anti-angiogenic combination therapy was confirmed. CONCLUSIONS Our study revealed that elevated PHLDA2 expression regulated by DNA hypomethylation was correlated with poor prognosis and immunosuppressive microenvironment, and highlighted the role of PHLDA2 as a robust biomarker for predicting therapeutic efficacy of ICI plus anti-angiogenic agent combination therapy in ccRCC, which expand the dimension of precision medicine.
Collapse
Affiliation(s)
- Junjie Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Xiuyi Pan
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Dingbang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Yu Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Xinyuan Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Chenhao Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Bo Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China.
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China.
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Xie W, Liu W, Wang L, Zhu B, Zhao C, Liao Z, Li Y, Jiang X, Liu J, Ren C. Roles of THEM4 in the Akt pathway: a double-edged sword. J Zhejiang Univ Sci B 2024; 25:541-556. [PMID: 39011675 PMCID: PMC11254685 DOI: 10.1631/jzus.b2300457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/10/2023] [Indexed: 07/17/2024]
Abstract
The protein kinase B (Akt) pathway can regulate the growth, proliferation, and metabolism of tumor cells and stem cells through the activation of multiple downstream target genes, thus affecting the development and treatment of a range of diseases. Thioesterase superfamily member 4 (THEM4), a member of the thioesterase superfamily, is one of the Akt kinase-binding proteins. Some studies on the mechanism of cancers and other diseases have shown that THEM4 binds to Akt to regulate its phosphorylation. Initially, THEM4 was considered an endogenous inhibitor of Akt, which can inhibit the phosphorylation of Akt in diseases such as lung cancer, pancreatic cancer, and liver cancer, but subsequently, THEM4 was shown to promote the proliferation of tumor cells by positively regulating Akt activity in breast cancer and nasopharyngeal carcinoma, which contradicts previous findings. Considering these two distinct views, this review summarizes the important roles of THEM4 in the Akt pathway, focusing on THEM4 as an Akt-binding protein and its regulatory relationship with Akt phosphorylation in various diseases, especially cancer. This work provides a better understanding of the roles of THEM4 combined with Akt in the treatment of diseases.
Collapse
Affiliation(s)
- Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Lei Wang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Bin Zhu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Cong Zhao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Hainan Province Clinical Medical Center, Sanya 572013, China. ,
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China.
| |
Collapse
|
23
|
Liu X, Xu W, Li L, Zhang Z, Lu M, Xia X. Dual PI3K/mTOR Inhibitor BEZ235 combined with BMS-1166 Promoting Apoptosis in Colorectal Cancer. Int J Med Sci 2024; 21:1814-1823. [PMID: 39113885 PMCID: PMC11302559 DOI: 10.7150/ijms.84320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
Background: BMS-1166, a PD-1/PD-L1 inhibitor, inhibits the binding of PD-L1 to PD-1, restores T cell function, and enhances tumor immune response. However, mutations in the tumor suppressor or impaired cellular signaling pathways may also lead to cellular transformation. In this study, the SW480 and SW480R cell lines were used as the model to elucidate the treatment with BMS-1166, BEZ235, and their combination. Methods: MTT and colony-formation assays were used to evaluate cell proliferation. Wound-healing assay was used to assess cell migration. Cell cycle and apoptosis were analyzed by flow cytometry. The phosphorylation level of the key kinases in the PI3K/Akt/mTOR and MAPK pathways, PD-L1, and the protein levels related to the proliferation, migration, and apoptosis were assessed using western blotting. Results: BEZ235 enhanced BMS-1166-mediated cell proliferation and migration inhibition in SW480 and SW480R cells and promoted apoptosis. Interestingly, the downregulation of the negative regulator PTEN raised the PD-L1 level, which was abolished by the inhibition of Akt. BMS-1166 promoted PI3K, Akt, mTOR, and Erk phosphorylation. However, the combination of BEZ235 with BMS-1166 suppressed the expression of PI3K, p-Akt, p-mTOR, and p-Erk in SW480 and SW480R cells compared to BMS-1166 or BEZ235 single treatment by inhibiting the binding of PD1 to PD-L1. Conclusions: PD-1 binds to PD-L1 and activates the PI3K/mTOR and MAPK pathways, which might be the molecular mechanism of acquired resistance of CRC to BMS-1166. The combination of the two drugs inhibited the phosphorylation of PI3K, Akt, and Erk in the PI3K/mTOR and MAPK pathway, i.e., BEZ235 enhanced the BMS-1166 treatment effect by blocking the PI3K/mTOR pathway and interfering with the crosstalk of the MAPK pathway. Therefore, these findings provide a theoretical basis for BMS-1166 combined with BEZ235 in the trial treatment of colorectal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoping Xia
- Department of Clinical Laboratory, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| |
Collapse
|
24
|
Wang L, Zhang X, Li H, Mou Y, Cui G. SP1 promotes high glucose-induced lens epithelial cell viability, migration and epithelial-mesenchymal transition via regulating FGF7 and PI3K/AKT pathway. Int Ophthalmol 2024; 44:316. [PMID: 38969958 DOI: 10.1007/s10792-024-03230-6] [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: 01/23/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Diabetic cataract (DC) is a common complication of diabetes and its etiology and progression are multi-factorial. In this study, the roles of specific protein 1 (SP1) and fibroblast growth factor 7 (FGF7) in DC development were explored. METHODS DC cell model was established by treating SRA01/04 cells with high glucose (HG). MTT assay was conducted to evaluate cell viability. Transwell assay and wound-healing assay were performed to assess cell migration and invasion. Western blot assay and qRT-PCR assay were conducted to measure the expression of N-cadherin, E-cadherin, Collagen I, Fibronectin, SP1 and FGF7 expression. CHIP assay and dual-luciferase reporter assay were conducted to analyze the combination between FGF7 and SP1. RESULTS FGF7 was upregulated in DC patients and HG-induced SRA01/04 cells. HG treatment promoted SRA01/04 cell viability, migration, invasion and epithelial-mesenchymal transition (EMT), while FGF7 knockdown abated the effects. Transcription factor SP1 activated the transcription level of FGF7 and SP1 overexpression aggravated HG-induced SRA01/04 cell injury. SP1 silencing repressed HG-induced SRA01/04 cell viability, migration, invasion and EMT, but these effects were ameliorated by upregulating FGF7. Additionally, SP1 knockdown inhibited the PI3K/AKT pathway by regulating the transcription level of FGF7. CONCLUSION Transcription factor SP1 activated the transcription level of FGF7 and the PI3K/AKT pathway to regulate HG-induced SRA01/04 cell viability, migration, invasion and EMT.
Collapse
Affiliation(s)
- Ledan Wang
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Xin Zhang
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Huijun Li
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Yuehong Mou
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Gangfeng Cui
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China.
| |
Collapse
|
25
|
Qi J, Wu Y, Guo Z, Zhu S, Xiong J, Hu F, Liang X, Ye X. Fibroblast growth factor 21 alleviates idiopathic pulmonary fibrosis by inhibiting PI3K-AKT-mTOR signaling and stimulating autophagy. Int J Biol Macromol 2024; 273:132896. [PMID: 38851619 DOI: 10.1016/j.ijbiomac.2024.132896] [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: 05/12/2023] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive pulmonary disease with an unclear pathogenesis and no available specific drug treatment. The principal etiological factors are lung inflammation caused by environmental factors, damage to alveolar epithelial cells, leading to epithelial-mesenchymal transition (EMT), and the abnormal proliferation of fibroblasts. Here, we have demonstrated that fibroblast growth factor 21 (FGF21) ameliorates IPF via the autophagy pathway. We administered FGF21 to bleomycin (BLM)-treated mice, which ameliorated their defects in lung function, reduced the accumulation of collagen, restored tissue structure, reduced the deposition of hydroxyproline, reduced the expression of collagen I and α-SMA and increased the expression of E-cadherin. The expression of LC3BII and the number of autophagosomes were significantly higher in the lungs. The expression of AKT and mTOR was significantly reduced by FGF21 treatment. We also determined the effects of FGF21 in A549 cells treated with TGF-β, and found that FGF21 significantly inhibits activation of the AKT signaling pathway, thereby reducing TGF-β-induced EMT and preventing the uncontrolled proliferation of fibroblasts. We conclude that FGF21 ameliorates IPF by inhibiting the PI3K-AKT-mTOR signaling pathway and activating autophagy, which provides a theoretical basis for FGF21 to be used for the treatment of IPF.
Collapse
Affiliation(s)
- Jianying Qi
- School of chemical engineering, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Yuanyuan Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Zhimou Guo
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jingjing Xiong
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Fei Hu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China.
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| |
Collapse
|
26
|
Liu M, Jiang H, Momeni MR. Epigenetic regulation of autophagy by non-coding RNAs and exosomal non-coding RNAs in colorectal cancer: A narrative review. Int J Biol Macromol 2024; 273:132732. [PMID: 38823748 DOI: 10.1016/j.ijbiomac.2024.132732] [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: 03/18/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
One of the major diseases affecting people globally is colorectal cancer (CRC), which is primarily caused by a lack of effective medical treatment and a limited understanding of its underlying mechanisms. Cellular autophagy functions to break down and eliminate superfluous proteins and substances, thereby facilitating the continual replacement of cellular elements and generating vital energy for cell processes. Non-coding RNAs and exosomal ncRNAs have a crucial impact on regulating gene expression and essential cellular functions such as autophagy, metastasis, and treatment resistance. The latest research has indicated that specific ncRNAs and exosomal ncRNA to influence the process of autophagy in CRC cells, which could have significant consequences for the advancement and treatment of this disease. It has been determined that a variety of ncRNAs have a vital function in regulating the genes essential for the formation and maturation of autophagosomes. Furthermore, it has been confirmed that ncRNAs have a considerable influence on the signaling pathways associated with autophagy, such as those involving AMPK, AKT, and mTOR. Additionally, numerous ncRNAs have the potential to affect specific genes involved in autophagy. This study delves into the control mechanisms of ncRNAs and exosomal ncRNAs and examines how they simultaneously influence autophagy in CRC.
Collapse
Affiliation(s)
- Minghua Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Hongfang Jiang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| | - Mohammad Reza Momeni
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
27
|
Hu J, Dai C, Ding Z, Pan Y, Lu L, Bao J, Zheng J. IKBIP promotes tumor development via the akt signaling pathway in esophageal squamous cell carcinoma. BMC Cancer 2024; 24:759. [PMID: 38914958 PMCID: PMC11197280 DOI: 10.1186/s12885-024-12510-4] [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: 02/20/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide. Inhibitor of kappa B kinase interacting protein (IKBIP) has been reported to promote glioma progression, but its role in other cancers remains unclear. This study aimed to investigate the role of IKBIP and its underlying molecular mechanisms in ESCC. METHODS The mRNA expression of IKBIP was analyzed using multiple cancer databases. Immunohistochemistry was performed to detect IKBIP protein expression in ESCC tissues and adjacent normal tissues, and Kaplan‒Meier survival and Cox regression analyses were carried out. The effects of IKBIP knockdown (or overexpression) on ESCC cells were detected by cell viability, cell migration, flow cytometry and Western blot assays. LY-294002 was used to validate the activation of the AKT signaling pathway by IKBIP. Finally, the role of IKBIP in ESCC was verified in a xenograft model. RESULTS Both bioinformatics analysis and immunohistochemistry indicated that IKBIP expression in ESCC tissues was significantly increased and was associated with the prognosis of ESCC patients. In vitro experiments revealed that IKBIP knockdown significantly inhibited the proliferation and migration of ESCC cells, and induced cell apoptosis and G1/S phase arrest. Molecular mechanism results showed that the AKT signaling pathway was further activated after IKBIP overexpression, thereby increasing the proliferation and migration abilities of ESCC cells. In vivo study confirmed that IKBIP promoted the initiation and development of ESCC tumors in mice. CONCLUSIONS IKBIP plays a tumor-promoting role in ESCC and may serve as a predictive biomarker and a potential therapeutic target for ESCC.
Collapse
Affiliation(s)
- Jiannan Hu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Chuanjing Dai
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Zhaoji Ding
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Yixiao Pan
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Lingxiao Lu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Jiaqian Bao
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China
| | - Jingmin Zheng
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximenjie, Linhai, Zhejiang Province, 317000, China.
| |
Collapse
|
28
|
He C, Wu Y, Nan X, Zhang W, Luo Y, Wang H, Li M, Liu C, Liu J, Mou X, Liu Y. Induction of CX3CL1 expression by LPS and its impact on invasion and migration in oral squamous cell carcinoma. Front Cell Dev Biol 2024; 12:1371323. [PMID: 38915444 PMCID: PMC11195639 DOI: 10.3389/fcell.2024.1371323] [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: 02/22/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
Purpose: This study aimed to explore the expression of CX3CL1 induced by lipopolysaccharide (LPS) in oral squamous cell carcinoma (OSCC) and its impact on biological characteristics such as invasion and migration, taking the foundation for new targets for the treatment and prognosis of OSCC. Methods: This study utilized a variety of techniques, including bioinformatics, molecular biology, and cell experiments, to investigate the expression of CX3CL1 and its receptor CX3CR1 in OSCC patients' cancer tissues or OSCC cell lines. Extracting, organizing, and analyzing the TCGA database on the expression of CX3CL1 and its receptor CX3CR1 in cancer tissues and corresponding paracancerous normal tissues of OSCC patients by bioinformatics methods. The expression of CX3CL1 in cancerous and normal tissues of OSCC patients was verified by IHC, and the changes in mRNA and protein expression of CX3CL1 and its receptor CX3CR1 in OSCC cell lines were detected before and after lipopolysaccharide LPS stimulation by RT-PCR, ELISA, and WB. Changes in cell biological behavior by overexpression of CX3CL1 in OSCC cell lines were detected by CCK-8, Transwell, scratch healing assay, and cloning assay. The effects of overexpressing cell lines on the AKT pathway and Epithelial-mesenchymal Transition (EMT)-related protein expression before and after LPS stimulation were detected by Western Blot. Results: (1) CX3CL1 and its receptor CX3CR1 were found to be downregulated in OSCC tissues of patients or OSCC cell lines. (2) After LPS stimulation, CX3CL1 gene expression increased in both OSCC cell lines, while CX3CR1 expression remained unchanged. (3) OSCC cell lines overexpressing CX3CL1 showed changes in cell biological characteristics, including decreased proliferation, invasion, migration, and stemness, which were more pronounced after LPS stimulation. (4) Overexpression of CX3CL1 in OSCC cell lines decreased EMT-related protein expression and AKT phosphorylation. On the contrary were promoted by LPS stimulation. Conclusion: CX3CL1 and CX3CR1 are downregulated in OSCC cancer tissues and cell lines compared to adjacent normal tissues and cells. LPS stimulation increases CX3CL1 expression in OSCC cell lines, suggesting that inflammation may induce CX3CL1 expression and that the CX3CL1 gene may play an important role in OSCC progression. Overexpression of CX3CL1 inhibits OSCC cell proliferation, migration, invasion, and stemness, suggesting that CX3CL1 plays a critical role in suppressing OSCC development. CX3CL1 suppresses OSCC invasion and migration by affecting EMT progression and AKT phosphorylation, and partially reverse the process that LPS causes and affects the development of OSCC.
Collapse
Affiliation(s)
- Chanjuan He
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Changsha Stomatological Hospital, Changsha, China
| | - Yuehan Wu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Xiaoxu Nan
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Weifang Zhang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Yu Luo
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Honglan Wang
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Mengqi Li
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Changyue Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Jiaming Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Xuelin Mou
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Ying Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
29
|
Wei Y, Yu P, Zhao Z, Sun C, Sun Q, Bai R, Gao W, Yang P. Cervical cancer cell-derived Tie1 expression via PI3K/AKT signaling pathway promotes tumor progression. Exp Cell Res 2024; 439:114060. [PMID: 38719173 DOI: 10.1016/j.yexcr.2024.114060] [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: 10/18/2023] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Tie1 orphan receptor has become a focus of research, Tie1 can form a polymer with Tie2, regulate the Ang/Tie2 pathway and play a vital role in pathological angiogenesis and tumor progression, the function of Tie1 has remained uncertain in the progression of cervical cancer (CC). Here, we investigated the functional influences of Tie1 overexpress on CC in vitro and in vivo. METHODS We used Immunohistochemistry (IHC) analysis to detect the relative expression of Tie1 in CC, and we analyzed its connection with the overall survival (OS) and progression free survival (PFS)of CC patients. To prove the role of Tie1 in cell proliferation and metastatic, Tie1 expression in CC cell lines was upregulated by lentivirus. RESULTS The high expression of Tie1 in tumor cells of cervical cancer tissues is significantly correlated with FIGO stage, differentiated tumors, tumors with diameters, deep stromal invasion. We found that cell progression was promoted in Tie1-overexpress CC cell lines in vivo and in vitro. Tie1 potentially exerts a commanding influence on the expression of markers associated with epithelial-mesenchymal transition (EMT) and the PI3K/AKT signaling pathway. CONCLUSIONS Our research indicates that Tie1 is highly connected to CC progression as it may play a role in the EMT process through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yan Wei
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Panpan Yu
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China; School of Medicine, Shihezi University, Shihezi, China
| | - Zouyu Zhao
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Chongfeng Sun
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Qianyu Sun
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Rui Bai
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Weirui Gao
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ping Yang
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| |
Collapse
|
30
|
Iqbal S, Karim MR, Mohammad S, Mathiyalagan R, Morshed MN, Yang DC, Bae H, Rupa EJ, Yang DU. Multiomics Analysis of the PHLDA Gene Family in Different Cancers and Their Clinical Prognostic Value. Curr Issues Mol Biol 2024; 46:5488-5510. [PMID: 38921000 PMCID: PMC11201736 DOI: 10.3390/cimb46060328] [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: 04/14/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
The PHLDA (pleckstrin homology-like domain family) gene family is popularly known as a potential biomarker for cancer identification, and members of the PHLDA family have become considered potentially viable targets for cancer treatments. The PHLDA gene family consists of PHLDA1, PHLDA2, and PHLDA3. The predictive significance of PHLDA genes in cancer remains unclear. To determine the role of pleckstrin as a prognostic biomarker in human cancers, we conducted a systematic multiomics investigation. Through various survival analyses, pleckstrin expression was evaluated, and their predictive significance in human tumors was discovered using a variety of online platforms. By analyzing the protein-protein interactions, we also chose a collection of well-known functional protein partners for pleckstrin. Investigations were also carried out on the relationship between pleckstrins and other cancers regarding mutations and copy number alterations. The cumulative impact of pleckstrin and their associated genes on various cancers, Gene Ontology (GO), and pathway analyses were used for their evaluation. Thus, the expression profiles of PHLDA family members and their prognosis in various cancers may be revealed by this study. During this multiomics analysis, we found that among the PHLDA family, PHLDA1 may be a therapeutic target for several cancers, including kidney, colon, and brain cancer, while PHLDA2 can be a therapeutic target for cancers of the colon, esophagus, and pancreas. Additionally, PHLDA3 may be a useful therapeutic target for ovarian, renal, and gastric cancer.
Collapse
Affiliation(s)
- Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (M.N.M.); (D.-C.Y.)
| | - Md. Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (M.N.M.); (D.-C.Y.)
| | - Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (R.M.)
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (R.M.)
| | - Md. Niaj Morshed
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (M.N.M.); (D.-C.Y.)
| | - Deok-Chun Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (M.N.M.); (D.-C.Y.)
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea;
| | - Hyocheol Bae
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea;
| | - Esrat Jahan Rupa
- College of Korean Medicine, Woosuk University, Wanju-gun 55338, Jeollabuk-do, Republic of Korea
| | - Dong Uk Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (M.N.M.); (D.-C.Y.)
| |
Collapse
|
31
|
FENG KUN, PENG HAO, LV QINGPENG, ZHANG YEWEI. PHLDA2 reshapes the immune microenvironment and induces drug resistance in hepatocellular carcinoma. Oncol Res 2024; 32:1063-1078. [PMID: 38827322 PMCID: PMC11136693 DOI: 10.32604/or.2024.047078] [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: 10/24/2023] [Accepted: 01/11/2024] [Indexed: 06/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignancy known for its unfavorable prognosis. The dysregulation of the tumor microenvironment (TME) can affect the sensitivity to immunotherapy or chemotherapy, leading to treatment failure. The elucidation of PHLDA2's involvement in HCC is imperative, and the clinical value of PHLDA2 is also underestimated. Here, bioinformatics analysis was performed in multiple cohorts to explore the phenotype and mechanism through which PHLDA2 may affect the progression of HCC. Then, the expression and function of PHLDA2 were examined via the qRT-PCR, Western Blot, and MTT assays. Our findings indicate a substantial upregulation of PHLDA2 in HCC, correlated with a poorer prognosis. The methylation levels of PHLDA2 were found to be lower in HCC tissues compared to normal liver tissues. Besides, noteworthy associations were observed between PHLDA2 expression and immune infiltration in HCC. In addition, PHLDA2 upregulation is closely associated with stemness features and immunotherapy or chemotherapy resistance in HCC. In vitro experiments showed that sorafenib or cisplatin significantly up-regulated PHLDA2 mRNA levels, and PHLDA2 knockdown markedly decreased the sensitivity of HCC cells to chemotherapy drugs. Meanwhile, we found that TGF-β induced the expression of PHLDA2 in vitro. The GSEA and in vitro experiment indicated that PHLDA2 may promote the HCC progression via activating the AKT signaling pathway. Our study revealed the novel role of PHLDA2 as an independent prognostic factor, which plays an essential role in TME remodeling and treatment resistance in HCC.
Collapse
Affiliation(s)
- KUN FENG
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - HAO PENG
- Medical School, Southeast University, Nanjing, 210000, China
| | - QINGPENG LV
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - YEWEI ZHANG
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| |
Collapse
|
32
|
Li Y, Zheng Y, Xu S, Hu H, Peng L, Zhu J, Wu M. The nanobody targeting PD-L1 and CXCR4 counteracts pancreatic stellate cell-mediated tumour progression by disrupting tumour microenvironment. Int Immunopharmacol 2024; 132:111944. [PMID: 38581990 DOI: 10.1016/j.intimp.2024.111944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/08/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal malignancy worldwide owing to its complex tumour microenvironment and dense physical barriers. Stromal-derived factor-1 (SDF-1), which is abundantly secreted by tumour stromal cells, plays a pivotal role in promoting PDAC growth and metastasis. In this study, we investigated the impact and molecular mechanisms of the anti-PD-L1&CXCR4 bispecific nanobody on the TME and their consequent interference with PDAC progression. We found that blocking the SDF-1/CXCR4 signalling pathway delayed the epithelial-mesenchymal transition in pancreatic cancer cells. Anti-PD-L1&CXCR4 bispecific nanobody effectively suppress the secretion of SDF-1 by pancreatic stellate cells and downregulate the expression of smooth muscle actin alpha(α-SMA), thereby preventing the activation of cancer-associated fibroblasts by downregulating the PI3K/AKT signaling pathway. This improves the pancreatic tumour microenvironment, favouring the infiltration of T cells into the tumour tissue. In conclusion, our results suggest that the anti-PD-L1&CXCR4 bispecific nanobody exerts an antitumor immune response by changing the pancreatic tumour microenvironment. Hence, the anti-PD-L1&CXCR4 bispecific nanobody is a potential candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Yaxian Li
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Yuejiang Zheng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Shuyi Xu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Hai Hu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Liyun Peng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Mingyuan Wu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| |
Collapse
|
33
|
Cai J, Yang Y, Zhang L, Fang Y, Zhang Y, Tan M, Zhang J, Tang C, Ren H, Wang L, Xiang G, Xu F, Lan L, Li L, Zheng X. Investigation of ENO2 as a promising novel marker for the progression of colorectal cancer with microsatellite instability-high. BMC Cancer 2024; 24:573. [PMID: 38724951 PMCID: PMC11080076 DOI: 10.1186/s12885-024-12332-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Microsatellite instability-high (MSI-H) has emerged as a significant biological characteristic of colorectal cancer (CRC). Studies reported that MSI-H CRC generally had a better prognosis than microsatellite stable (MSS)/microsatellite instability-low (MSI-L) CRC, but some MSI-H CRC patients exhibited distinctive molecular characteristics and experienced a less favorable prognosis. In this study, our objective was to explore the metabolic transcript-related subtypes of MSI-H CRC and identify a biomarker for predicting survival outcomes. METHODS Single-cell RNA sequencing (scRNA-seq) data of MSI-H CRC patients were obtained from the Gene Expression Omnibus (GEO) database. By utilizing the copy number variation (CNV) score, a malignant cell subpopulation was identified at the single-cell level. The metabolic landscape of various cell types was examined using metabolic pathway gene sets. Subsequently, functional experiments were conducted to investigate the biological significance of the hub gene in MSI-H CRC. Finally, the predictive potential of the hub gene was assessed using a nomogram. RESULTS This study revealed a malignant tumor cell subpopulation from the single-cell RNA sequencing (scRNA-seq) data. MSI-H CRC was clustered into two subtypes based on the expression profiles of metabolism-related genes, and ENO2 was identified as a hub gene. Functional experiments with ENO2 knockdown and overexpression demonstrated its role in promoting CRC cell migration, invasion, glycolysis, and epithelial-mesenchymal transition (EMT) in vitro. High expression of ENO2 in MSI-H CRC patients was associated with worse clinical outcomes, including increased tumor invasion depth (p = 0.007) and greater likelihood of perineural invasion (p = 0.015). Furthermore, the nomogram and calibration curves based on ENO2 showed potential prognosis predictive performance. CONCLUSION Our findings suggest that ENO2 serves as a novel prognostic biomarker and is associated with the progression of MSI-H CRC.
Collapse
Affiliation(s)
- Junwen Cai
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Yuting Yang
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Leilei Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Yangyang Fang
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Yanjun Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Mingyue Tan
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Juan Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Chen Tang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Haitao Ren
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Lanni Wang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Guangxin Xiang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Feng Xu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Liyi Li
- General Surgery Department, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoqun Zheng
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
- The Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, China.
| |
Collapse
|
34
|
Zhang X, Zhu R, Jiao Y, Simayi H, He J, Shen Z, Wang H, He J, Zhang S, Yang F. Expression profiles and gene set enrichment analysis of the transcriptomes from the cancer tissue, white adipose tissue and paracancer tissue with colorectal cancer. PeerJ 2024; 12:e17105. [PMID: 38563016 PMCID: PMC10984182 DOI: 10.7717/peerj.17105] [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: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common cancers worldwide and is related to diet and obesity. Currently, crosstalk between lipid metabolism and CRC has been reported; however, the specific mechanism is not yet understood. In this study, we screened differentially expressed long non-coding RNAs (lncRNAs) and mRNAs from primary cancer, paracancer, and white adipose tissue of CRC patients. We screened and analyzed the genes differentially expressed between primary and paracancer tissue and between paracancer and white adipose tissue but not between primary and white adipose tissue. According to the results of the biological analysis, we speculated a lncRNA (MIR503HG) that may be involved in the crosstalk between CRC and lipid metabolism through exosome delivery. Methods We screened differentially expressed long non-coding RNAs (lncRNAs) and mRNAs from primary cancer, paracancer, and white adipose tissue of CRC patients. We screened and analyzed the genes differentially expressed between primary and paracancer tissue and between paracancer and white adipose tissue but not between primary and white adipose tissue. Results We speculated a lncRNA (MIR503HG) that may be involved in the crosstalk between CRC and lipid metabolism through exosome delivery. Conclusions In this study, the findings raise the possibility of crosstalk between lipid metabolism and CRC through the exosomal delivery of lncRNAs.
Collapse
Affiliation(s)
- Xiufeng Zhang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine; Zhejiang Provincial Clinical Research Center, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Rui Zhu
- Affiliated XiaoShan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Ye Jiao
- Chronic Disease Research Institute, The Children’s Hospital, and National Clinical Research Center for Child Health, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Halizere Simayi
- Chronic Disease Research Institute, The Children’s Hospital, and National Clinical Research Center for Child Health, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jialing He
- Department of Colorectal Surgery, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhong Shen
- Department of Colorectal Surgery, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Houdong Wang
- Department of Colorectal Surgery, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun He
- Department of Colorectal Surgery, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Suzhan Zhang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine; Zhejiang Provincial Clinical Research Center, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei Yang
- Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Simoni MK, Negatu SG, Park JY, Mani S, Arreguin MC, Amses K, Huh DD, Mainigi M, Jurado KA. Type I interferon alters invasive extravillous trophoblast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584521. [PMID: 38559122 PMCID: PMC10979977 DOI: 10.1101/2024.03.11.584521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Inappropriate type I interferon (IFN) signaling during embryo implantation and placentation is linked to poor pregnancy outcomes. Here, we evaluated the consequence of elevated type I IFN exposure on implantation using a biomimetic model of human implantation in an organ-on-a-chip device. We found that type I IFN reduced extravillous trophoblast (EVT) invasion capacity. Analyzing single-cell transcriptomes, we uncovered that IFN truncated endovascular EVT emergence in the implantation-on-a-chip device by stunting EVT epithelial-to-mesenchymal transition. Disruptions to the epithelial-to-mesenchymal transition is associated with the pathogenesis of preeclampsia, a life-threatening hypertensive disorder of pregnancy. Strikingly, unwarranted IFN stimulation induced genes associated with increased preeclampsia risk and a preeclamptic gene-like signature in EVTs. These dysregulated EVT phenotypes ultimately reduced EVT-mediated endothelial cell vascular remodeling in the implantation-on-a-chip device. Overall, our work indicates IFN signaling can alter EVT epithelial-to-mesenchymal transition progression which results in diminished EVT-mediated spiral artery remodeling and a preeclampsia gene signature upon sustained stimulation. Our work implicates unwarranted type I IFN as a maternal disturbance that can result in abnormal EVT function that could trigger preeclampsia.
Collapse
|
36
|
Li C, Cao S, Guo M, Guo A, Sun X. Identification of potential key genes for colorectal cancer based on bioinformatics analysis. Medicine (Baltimore) 2023; 102:e36615. [PMID: 38134110 PMCID: PMC10735105 DOI: 10.1097/md.0000000000036615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
This study aimed to explore key genes as potential biomarkers for colorectal cancer (CRC) diagnosis and prognosis in order to improve their clinical utility. To identify and screen candidate genes involved in CRC carcinogenesis and disease progression, we downloaded the microarray datasets GSE143939, GSE196006, and GSE200427 from the GEO database and applied the GEO2R tool to obtain differentially expressed genes (DEGs) between colorectal cancer tissue samples and normal tissue samples. Differentially expressed genes were analyzed using the DAVID online database for gene ontology and Kyoto encyclopedia of genes and genomes pathway enrichment analyses. Protein-protein interaction network was constructed and related module analysis was performed using STRING and Cytoscape. In total, 241 DEGs were identified, including 127 downregulated and 114 upregulated genes. DEGs enriched functions and pathways included cellular response to chemical stimulus, extracellular region, carbonate dehydratase activity, cell division, spindle, and cell division. The abundant functions and pathways of DEGs included cellular response to chemical stimulus, extracellular region, carbonate dehydratase activity, cell division, spindle, cell adhesion molecule binding, Aldosterone-regulated sodium reabsorption, and Cell cycle-related processes. Fifteen key genes were identified, and bioprocess analyses showed that these genes were mainly enriched in cell cycle, cell division, mitotic spindle, and tubulin binding processes. It was found that CDK1, CEP55, MKI67, and TOP2A may be involved in CRC cancer invasion and recurrence. The pivotal genes identified in this study contribute to our understanding of the molecular and pathogenic mechanisms of CRC carcinogenesis and progression, and provide possible biomarkers for the diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Chongyang Li
- Second Clinical Medical College, Binzhou Medical University, Yantai, China
- Jinan Fourth People’s Hospital, Jinan, China
| | | | - Mingxiao Guo
- Department of General Surgery Center, Linyi People’s Hospital, Linyi, China
| | - Aihong Guo
- Jinan Fourth People’s Hospital, Jinan, China
| | - Xuedi Sun
- Jinan Fourth People’s Hospital, Jinan, China
- Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
37
|
Liu H, Guo H, Wu Y, Hu Q, Hu G, He H, Yin Y, Nan X, Lin G, Han J, Zhao R, Liu Y. RCN1 deficiency inhibits oral squamous cell carcinoma progression and THP-1 macrophage M2 polarization. Sci Rep 2023; 13:21488. [PMID: 38057406 PMCID: PMC10700561 DOI: 10.1038/s41598-023-48801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
Reticulocalbin 1 (RCN1), a calcium-binding protein located in the endoplasmic reticulum (ER) lumen, contains six conserved regions. Its main functions include maintaining intracellular homeostasis and regulating cell proliferation and apoptosis, and it plays an important role in the development of various tumours. However, the exact function of RCN1 in oral squamous cell carcinoma (OSCC) is not fully understood. Therefore, the aim of this study was to investigate the effects of RCN1 on the biological behaviour of OSCC and the regulation of tumour-associated macrophage (TAM) polarization. The expression of RCN1 in OSCC and normal oral mucosa was evaluated through bioinformatics analysis and immunohistochemical staining. The growth, migration, and invasion of OSCC cells were observed after knockdown of RCN1 using CCK-8 and Transwell assays. Apoptosis was detected by flow cytometry. The effect of tumour cell-derived RCN1 on the polarization of THP-1 macrophages was investigated by establishing a coculture model of THP-1 macrophages and OSCC cells. Additionally, changes in the expression levels of relevant proteins were detected using Western blotting. The upregulation of RCN1 in tumour tissues compared to normal oral mucosal tissues is associated with a poor prognosis and can be utilized as a prognostic indicator for OSCC. Knockdown of RCN1 inhibited the proliferation, migration, and invasion of OSCC cells. Additionally, knockdown of RCN1 in Cal-27 and SCC-25 cells resulted in inhibition of the M2 polarization of THP-1 macrophages. RCN1 knockdown inhibits OSCC progression and M2 macrophage polarization. Targeting RCN1 may be a promising approach for OSCC treatment.
Collapse
Affiliation(s)
- Han Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Haiyang Guo
- Digestive Endoscopy Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yuehan Wu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Qiannan Hu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Guangbing Hu
- Institute of Hepato-Biliary-Pancreatic-Intestinal Disease, North Sichuan Medical College, Nanchong, China
| | - Huan He
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yaolin Yin
- Institute of Hepato-Biliary-Pancreatic-Intestinal Disease, North Sichuan Medical College, Nanchong, China
| | - Xiaoxu Nan
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Gaoren Lin
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Jinpeng Han
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Runzhe Zhao
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Ying Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
- Department of Stomatology, North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
38
|
Wang S, Wang YF, Yang G, Zhang HH, Yuan HF, Hou CY, Zhao LN, Suo YH, Sun J, Sun LL, Lv P, Sun Y, Zhang NN, Zhang XD, Lu W. Heat shock protein family A member 8 serving as a co-activator of transcriptional factor ETV4 up-regulates PHLDA2 to promote the growth of liver cancer. Acta Pharmacol Sin 2023; 44:2525-2536. [PMID: 37474643 PMCID: PMC10692233 DOI: 10.1038/s41401-023-01133-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
Heat shock protein family A member 8 (HSPA8) participates in the folding or degradation of misfolded proteins under stress and plays critical roles in cancer. In this study, we investigated the function of HSPA8 in the development of liver cancer. By analyzing the TCGA transcriptome dataset, we found that HSPA8 was upregulated in 134 clinical liver cancer tissue samples, and positively correlated with poor prognosis. IHC staining showed the nuclear and cytoplasmic localization of HSPA8 in liver cancer cells. Knockdown of HSPA8 resulted in a decrease in the proliferation of HepG2 and Huh-7 cells. ChIP-seq and RNA-seq analysis revealed that HSPA8 bound to the promoter of pleckstrin homology-like domain family A member 2 (PHLDA2) and regulated its expression. The transcription factor ETV4 in HepG2 cells activated PHLDA2 transcription. HSPA8 and ETV4 could interact with each other in the cells and colocalize in the nucleus. From a functional perspective, we demonstrated that HSPA8 upregulated PHDLA2 through the coactivating transcription factor ETV4 to enhance the growth of liver cancer in vitro and in vivo. From a therapeutic perspective, we identified both HSPA8 and PHDLA2 as novel targets in the treatment of HCC. In conclusion, this study demonstrates that HSPA8 serves as a coactivator of ETV4 and upregulates PHLDA2, leading to the growth of HCC, and is a potential therapeutic target in HCC treatment.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Yu-Fei Wang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Guang Yang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hui-Hui Zhang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hong-Feng Yuan
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chun-Yu Hou
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li-Na Zhao
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yu-Hong Suo
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Jiao Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Lin-Lin Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Pan Lv
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yan Sun
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China.
| | - Ning-Ning Zhang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China.
| | - Xiao-Dong Zhang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Wei Lu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
39
|
Deris Zayeri Z, Parsi A, Shahrabi S, Kargar M, Davari N, Saki N. Epigenetic and metabolic reprogramming in inflammatory bowel diseases: diagnostic and prognostic biomarkers in colorectal cancer. Cancer Cell Int 2023; 23:264. [PMID: 37936149 PMCID: PMC10631091 DOI: 10.1186/s12935-023-03117-z] [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: 05/06/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND AND AIM "Inflammatory bowel disease" (IBD) is a chronic, relapsing inflammatory disease of the intestinal tract that typically begins at a young age and might transit to colorectal cancer (CRC). In this manuscript, we discussed the epigenetic and metabolic change to present a extensive view of IBDs transition to CRC. This study discusses the possible biomarkers for evaluating the condition of IBDs patients, especially before the transition to CRC. RESEARCH APPROACH We searched "PubMed" and "Google Scholar" using the keywords from 2000 to 2022. DISCUSSION In this manuscript, interesting titles associated with IBD and CRC are discussed to present a broad view regarding the epigenetic and metabolic reprogramming and the biomarkers. CONCLUSION Epigenetics can be the main reason in IBD transition to CRC, and Hypermethylation of several genes, such as VIM, OSM4, SEPT9, GATA4 and GATA5, NDRG4, BMP3, ITGA4 and plus hypomethylation of LINE1 can be used in IBD and CRC management. Epigenetic, metabolisms and microbiome-derived biomarkers, such as Linoleic acid and 12 hydroxy 8,10-octadecadienoic acid, Serum M2-pyruvate kinase and Six metabolic genes (NAT2, XDH, GPX3, AKR1C4, SPHK and ADCY5) expression are valuable biomarkers for early detection and transition to CRC condition. Some miRs, such as miR-31, miR-139-5p, miR -155, miR-17, miR-223, miR-370-3p, miR-31, miR -106a, miR -135b and miR-320 can be used as biomarkers to estimate IBD transition to CRC condition.
Collapse
Affiliation(s)
- Zeinab Deris Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abazar Parsi
- Alimentary Tract Research Center, Clinical Sciences Research Inistitute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Masoud Kargar
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
40
|
YANG X, WANG J, CHENG L, ZHANG Y, HUANG J, LIU M. Active compounds of Caodoukou () inhibit the migration, invasion and metastasis of human pancreatic cancer cells by targeting phosphoinosmde-3-kinase/ protein kinase B/mammalian target of rapamycin pathway. J TRADIT CHIN MED 2023; 43:876-886. [PMID: 37679975 PMCID: PMC10465845 DOI: 10.19852/j.cnki.jtcm.20230802.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/19/2022] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To detect the effects of active compounds of Caodoukou () (ACAK) on the proliferation, migration and invasion of pancreatic cancer, and explain the possible molecular mechanism of ACAK interacting with these processes. METHODS Cell counting kit-8 method, cell scratch repair experiment, Transwell migration and invasion experiment, immunohistochemistry, western blot assay and real-time polymerase chain reaction experiment were used to evaluate the effect of ACAK on the proliferation, migration and invasion of pancreatic cancer cells. The levels of active molecules involved in the phosphoinosmde-3-kinase (PI3K)/Akt/the mammalian target of rapamycin (mTOR) signal transduction were detected by Western blot assay. In addition, the function of ACAK was evaluated by xenotransplantation tumor model in nude mice. RESULTS The inhibitory effect of ACAK on the proliferation of pancreatic cancer cells showed certain time-dose dependence. The results of scratch repair test, Transwell test, Western blotting and real time polymerase chain reaction assay showed that ACAK could inhibit the migration and invasion of pancreatic cancer cells . In addition, the regulatory effect of ACAK on epithelial-mesenchymal transition (EMT) is partly attributed to PI3K/Akt/mTOR signaling pathway. The experimental results showed that ACAK regulated the development of pancreatic cancer. CONCLUSIONS ACAK can partly inhibit the activity of EMT and matrix metallopeptidases by down-regulating the downstream proteins of PI3K/Akt/mTOR signal pathway, thus inhibiting the ability of migration and invasion of pancreatic cancer.
Collapse
Affiliation(s)
- Xiaohui YANG
- 1 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jian WANG
- 1 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Li CHENG
- 1 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yuxi ZHANG
- 1 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jianlin HUANG
- 2 Department of Pharmacy, Luzhou Naxi District People's Hospital, Luzhou 646000, China
- 3 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Minghua LIU
- 1 Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
41
|
Chakraborty S, Coleman C, Manoj P, Demircioglu D, Shah N, de Stanchina E, Rudin CM, Hasson D, Sen T. De Novo and Histologically Transformed Small-Cell Lung Cancer Is Sensitive to Lurbinectedin Treatment Through the Modulation of EMT and NOTCH Signaling Pathways. Clin Cancer Res 2023; 29:3526-3540. [PMID: 37382635 PMCID: PMC10901109 DOI: 10.1158/1078-0432.ccr-23-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 06/27/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE Small-cell lung cancer (SCLC) is a high-grade neuroendocrine tumor with dismal prognosis and limited treatment options. Lurbinectedin, conditionally approved as a second-line treatment for metastatic SCLC, drives clinical responses in about 35% of patients, and the overall survival (OS) of those who benefit from it remains very low (∼9.3 months). This finding highlights the need to develop improved mechanistic insight and predictive biomarkers of response. EXPERIMENTAL DESIGN We used human and patient-derived xenograft (PDX)-derived SCLC cell lines to evaluate the effect of lurbinectedin in vitro. We also demonstrate the antitumor effect of lurbinectedin in multiple de novo and transformed SCLC PDX models. Changes in gene and protein expression pre- and post-lurbinectedin treatment was assessed by RNA sequencing and Western blot analysis. RESULTS Lurbinectedin markedly reduced cell viability in the majority of SCLC models with the best response on POU2F3-driven SCLC cells. We further demonstrate that lurbinectedin, either as a single agent or in combination with osimertinib, causes an appreciable antitumor response in multiple models of EGFR-mutant lung adenocarcinoma with histologic transformation to SCLC. Transcriptomic analysis identified induction of apoptosis, repression of epithelial-mesenchymal transition, modulation of PI3K/AKT, NOTCH signaling associated with lurbinectedin response in de novo, and transformed SCLC models. CONCLUSIONS Our study provides a mechanistic insight into lurbinectedin response in SCLC and the first demonstration that lurbinectedin is a potential therapeutic target after SCLC transformation.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Charles Coleman
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Deniz Demircioglu
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nisargbhai Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dan Hasson
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Mount Sinai, New York, New York
| |
Collapse
|
42
|
Yue L, Xu X, Dai S, Xu F, Zhao W, Gu J, Dai X, Qian X. Orosomucoid 1 promotes colorectal cancer progression and liver metastasis by affecting PI3K/AKT pathway and inducing macrophage M2 polarization. Sci Rep 2023; 13:14092. [PMID: 37640741 PMCID: PMC10462626 DOI: 10.1038/s41598-023-40404-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Approximately 25-30% of those affected by colorectal cancer (CRC), the most prevalent gastrointestinal malignancy, develop metastases. The survival rate of patients with liver metastasis of CRC (CRLM) remains low owing to its unpredictability and a lack of biomarkers that can be applied to distinguish groups at higher risk for CRLM among patients with CRC. Therefore, our study aimed to find biomarkers that can predict the risk of CRLM. Screening of the Gene Expression Omnibus database, supported by an analysis of clinically obtained tissue and serum data using qPCR and ELISA, in an attempt to identify relevant biomarkers, enabled us to determine that orosomucoid 1 (ORM1) was differentially expressed in liver metastases and primary tumors of patients with CRC. Functionally, overexpression of ORM1 promoted the epithelial-mesenchymal transition and the proliferative, migratory, and invasive activities of MC38 cells and activated the PI3K/AKT signaling pathway. Moreover, MC38 cells overexpressing ORM1 enhanced the tumor immune microenvironment by promoting macrophage M2 polarization and elevating interleukin-10 (IL-10) expression. In vivo experiments further confirmed in vitro results, indicating that liver metastases elevated by ORM1 were partially attenuated by the depletion of macrophages or IL-10. Considered together, ORM1 promotes CRC progression and liver metastasis by regulating tumor cell growth and inducing macrophage M2 polarization, which mediates tumor immune tolerance, and thus acts as a potential predictive marker and therapeutic target in CRLM.
Collapse
Affiliation(s)
- Lei Yue
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xiaozhang Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Shipeng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Fan Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wenhu Zhao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Xinzheng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China.
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| | - Xiaofeng Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, Jiangsu Province, China.
- NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
43
|
Zhou C, Huang Y, Nie S, Zhou S, Gao X, Chen G. Biological effects and mechanisms of fisetin in cancer: a promising anti-cancer agent. Eur J Med Res 2023; 28:297. [PMID: 37626424 PMCID: PMC10464434 DOI: 10.1186/s40001-023-01271-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Fisetin, a natural flavonoid, possesses numerous biological activities that have been extensively studied in various diseases. When it comes to cancer, fisetin exhibits a range of biological effects, such as suppressing cell growth, triggering programmed cell death, reducing the formation of new blood vessels, protecting against oxidative stress, and inhibiting cell migration. Moreover, fisetin has the ability to enhance the effectiveness of chemotherapy. The anticancer properties of fisetin can be attributed to a diverse array of molecules and signaling pathways, including vascular endothelial growth factor (VEGF), mitogen-activated protein kinase (MAPK), nuclear factor-kappa B (NF-κB), PI3K/Akt/mTOR, and Nrf2/HO-1. Consequently, fisetin holds promise as a therapeutic agent for anticancer treatment. In this review, we place emphasis on the biological functions and various molecular targets of fisetin in anticancer therapy.
Collapse
Affiliation(s)
- Chenhui Zhou
- School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Yi Huang
- School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Sheng Nie
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Shengjun Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Xiang Gao
- School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China.
| | - Gao Chen
- School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
44
|
Shi X, Li H, Dan Z, Shu C, Zhu R, Yang Q, Wang Y, Zhu H. Melatonin Potentiates Sensitivity to 5-Fluorouracil in Gastric Cancer Cells by Upregulating Autophagy and Downregulating Myosin Light-Chain Kinase. J Cancer 2023; 14:2608-2618. [PMID: 37779875 PMCID: PMC10539390 DOI: 10.7150/jca.85353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/23/2023] [Indexed: 10/03/2023] Open
Abstract
5-Fluorouracil is an effective chemotherapeutic drug for gastric cancer. However, the acquisition of chemotherapeutic resistance remains a challenge in treatment. Melatonin can enhance the therapeutic effect of 5-fluorouracil; however, the underlying mechanisms are not well understood. We investigated the effects of combinations of melatonin and 5-fluorouracil on the proliferation, migration and invasion of gastric cancer cells. Melatonin significantly potentiated the 5-fluorouracil-mediated inhibition of proliferation, migration and invasion in gastric cancer cells, which potentiates sensitivity to 5-FU by promoting the activation of Beclin-1-dependent autophagy and targeting the myosin light-chain kinase (MLCK) signaling pathway. Previous studies have shown that autophagy might be associated with the MLCK signaling pathway. The autophagy inhibitor, 3-methyladenine, effectively rescued the migratory and invasive capabilities of gastric cancer cells, while also reducing expression level of MLCK and the phosphorylation level of MLC. This indicates that autophagy is involved in tumor metastasis, which may be related to inhibition of the MLCK signaling pathway. Our findings indicate that melatonin can improve the effectiveness of 5-fluorouracil in gastric cancer and could be used as a supplemental agent in the treatment of gastric cancer with 5-fluorouracil.
Collapse
Affiliation(s)
- Xiaorui Shi
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhangyong Dan
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Chuanlin Shu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Rumeng Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Qingling Yang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Yi Wang
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
- Department of Biological Engineering, School of Life Sciences, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Huaqing Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
45
|
Yan YC, Meng GX, Yang CC, Yang YF, Tan SY, Yan LJ, Ding ZN, Ma YL, Dong ZR, Li T. Diacylglycerol lipase alpha promotes hepatocellular carcinoma progression and induces lenvatinib resistance by enhancing YAP activity. Cell Death Dis 2023; 14:404. [PMID: 37414748 PMCID: PMC10325985 DOI: 10.1038/s41419-023-05919-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023]
Abstract
As an important hydrolytic enzyme that yields 2-AG and free fatty acids, diacylglycerol lipase alpha (DAGLA) is involved in exacerbating malignant phenotypes and cancer progression, but the role of the DAGLA/2-AG axis in HCC progression remains unclear. Here, we found that the upregulation of components of the DAGLA/2-AG axis in HCC samples is correlated with tumour stage and patient prognosis. In vitro and in vivo experiments demonstrated that the DAGLA/2-AG axis promoted HCC progression by regulating cell proliferation, invasion and metastasis. Mechanistically, the DAGLA/2AG axis significantly inhibited LATS1 and YAP phosphorylation, promoted YAP nuclear translocation and activity, and ultimately led to TEAD2 upregulation and increased PHLDA2 expression, which could be enhanced by DAGLA/2AG-induced activation of the PI3K/AKT pathway. More importantly, DAGLA induced resistance to lenvatinib therapy during HCC treatment. Our study demonstrates that inhibiting the DAGLA/2-AG axis could be a novel therapeutic strategy to inhibit HCC progression and enhance the therapeutic effects of TKIs, which warrant further clinical studies.
Collapse
Affiliation(s)
- Yu-Chuan Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Guang-Xiao Meng
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Chun-Cheng Yang
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Ya-Fei Yang
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, 250012, Jinan, China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Yun-Long Ma
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China.
| |
Collapse
|
46
|
Ma TF, Fan YR, Zhao YH, Liu B. Emerging role of autophagy in colorectal cancer: Progress and prospects for clinical intervention. World J Gastrointest Oncol 2023; 15:979-987. [PMID: 37389106 PMCID: PMC10302997 DOI: 10.4251/wjgo.v15.i6.979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 06/14/2023] Open
Abstract
Autophagy is a physiological mechanism in which cells degrade themselves and quickly recover the degraded cell components. Recent studies have shown that autophagy plays an important role in the occurrence, development, treatment, and prognosis of colorectal cancer. In the early stages of colorectal cancer, autophagy can inhibit the production and development of tumors through multiple mechanisms such as maintaining DNA stability, inducing tumor death, and enhancing immune surveillance. However, as colorectal cancer progresses, autophagy may mediate tumor resistance, enhance tumor metabolism, and other pathways to promote tumor development. Therefore, intervening in autophagy at the appropriate time has broad clinical application prospects. This article summarizes the recent research progress of autophagy and colorectal cancer and is expected to provide new theoretical basis and reference for clinical treatment of colorectal cancer.
Collapse
Affiliation(s)
- Tian-Fei Ma
- Department of Breast Internal Medicine Ⅱ, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Yue-Ren Fan
- Department of Breast Internal Medicine Ⅱ, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Yi-Hang Zhao
- Department of Breast Internal Medicine Ⅱ, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Bin Liu
- Department of Breast Internal Medicine Ⅱ, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
47
|
Jin C, Wang T, Yang Y, Zhou P, Li J, Wu W, Lv X, Ma G, Wang A. Rational targeting of autophagy in colorectal cancer therapy: From molecular interactions to pharmacological compounds. ENVIRONMENTAL RESEARCH 2023; 227:115721. [PMID: 36965788 DOI: 10.1016/j.envres.2023.115721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
The abnormal progression of tumors has been a problem for treatment of cancer and therapeutic should be directed towards targeting main mechanisms involved in tumorigenesis in tumors. The genomic mutations can result in changes in biological mechanisms in human cancers. Colorectal cancer is one of the most malignant tumors of gastrointestinal tract and its treatment has been faced some difficulties due to development of resistance in tumor cells and also, their malignant behavior. Hence, new therapeutic modalities for colorectal cancer are being investigated. Autophagy is a "self-digestion" mechanism that is responsible for homeostasis preserving in cells and its aberrant activation/inhibition can lead to tumorigenesis. The current review focuses on the role of autophagy mechanism in colorectal cancer. Autophagy may be associated with increase/decrease in progression of colorectal cancer due to mutual function of this molecular mechanism. Pro-survival autophagy inhibits apoptosis to increase proliferation and survival rate of colorectal tumor cells and it is also involved in cancer metastasis maybe due to EMT induction. In contrast, pro-death autophagy decreases growth and invasion of colorectal tumor cells. The status of autophagy (upregulation and down-regulation) is a determining factor for therapy response in colorectal tumor cells. Therefore, targeting autophagy can increase sensitivity of colorectal tumor cells to chemotherapy and radiotherapy. Interestingly, nanoparticles can be employed for targeting autophagy in cancer therapy and they can both induce/suppress autophagy in tumor cells. Furthermore, autophagy modulators can be embedded in nanostructures in improving tumor suppression and providing cancer immunotherapy.
Collapse
Affiliation(s)
- Canhui Jin
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Tianbao Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Yanhui Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhou
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Juncheng Li
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Wenhao Wu
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Xin Lv
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Guoqing Ma
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Aihong Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China.
| |
Collapse
|
48
|
Liu P, Li L, Wang W, He C, Xu C. MST4 promotes proliferation, invasion, and metastasis of gastric cancer by enhancing autophagy. Heliyon 2023; 9:e16735. [PMID: 37313160 PMCID: PMC10258413 DOI: 10.1016/j.heliyon.2023.e16735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Background Mammalian infertile-20-like kinase 4 (MST4) plays major roles in the progression of malignant tumor types, but its function in gastric cancer (GC) remains poorly understood. Objective To investigate the regulatory mechanism of MST4 in GC. Methods Immunohistochemistry was used to detect MST4 protein in GC tissue. Additionally, the correlation between MST4 expression and the clinicopathological characteristics and prognosis of GC was evaluated. The MST4 expression level in GC cells was measured by western blotting and quantitative real-time polymerase chain reaction. Moreover, the regulatory mechanism of MST4 was investigated in vitro and in vivo. Results Overexpression of MST4 was found in GC tissue and cell lines, which correlated to the tumor size, histological type, invasion depth, ulcer, lymph node metastasis, lymphovascular invasion, perineural invasion and TNM stage (all P < 0.01). In terms of MST4 functions in vitro, its upregulation facilitated the proliferation, migration, and invasion of GC cells. Furthermore, MST4 promoted these processes by facilitating autophagy, whereas downregulation of MST4 significantly attenuated these processes. Downregulation of MST4 also attenuated tumor growth in vivo. Conclusion High expression of MST4 indicates a poor prognosis and promotes GC cell proliferation, invasion, and metastasis by enhancing autophagy.
Collapse
Affiliation(s)
- Pengwei Liu
- Departments of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Departments of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Lin Li
- Departments of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Wei Wang
- Departments of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Chiyi He
- Departments of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Chunfang Xu
- Departments of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| |
Collapse
|
49
|
Liu G, Wang H, Ran R, Wang Y, Li Y. FOSL1 transcriptionally regulates PHLDA2 to promote 5-FU resistance in colon cancer cells. Pathol Res Pract 2023; 246:154496. [PMID: 37178619 DOI: 10.1016/j.prp.2023.154496] [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: 11/08/2022] [Revised: 04/11/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Tumor drug resistance is a leading cause of tumor treatment failure. To date, the association between FOS-Like antigen-1 (FOSL1) and chemotherapy sensitivity in colon cancer is unclear. The present study investigated the molecular mechanism of FOSL1 regulating 5-Fluorouracil (5-FU) resistance in colon cancer. METHODS FOSL1 expression in colon cancer was analyzed by bioinformatics methods, and its downstream regulatory factors were predicted. Pearson correlation analyzed the expression of FOSL1 and downstream regulatory gene. Meanwhile, the expression of FOSL1 and its downstream factor Pleckstrin Homology-Like Domain Family A Member 2 (PHLDA2) in colon cancer cell lines was measured by qRT-PCR and western blot. The regulatory relationship between FOSL1 and PHLDA2 was verified by chromatin immunoprecipitation (ChIP) assay and dual-luciferase reporter assay. The effects of the FOSL1/PHLDA2 axis on the resistance in colon cancer cells to 5-FU were analyzed by cell experiments. RESULTS FOSL1 expression was evidently up-regulated in colon cancer and 5-FU resistant cells. FOSL1 was positively correlated with PHLDA2 in colon cancer. In vitro cell assays showed that low expression of FOSL1 significantly enhanced 5-FU sensitivity in colon cancer cells, significantly suppressed the proliferation of cancer cells, and induced apoptosis. Overexpression of FOSL1 presented the opposite regulatory trend. Mechanistically, FOSL1 activated PHLDA2 and up-regulated its expression. Moreover, by activating glycolysis, PHLDA2 promoted 5-Fu resistance and cell proliferation, and reduced cell apoptosis in colon cancer. CONCLUSION Down-regulated FOSL1 expression could enhance the 5-FU sensitivity of colon cancer cells, and FOSL1/PHLDA2 axis may be an effective target for overcoming chemotherapy resistance in colon cancer.
Collapse
Affiliation(s)
- Guangyi Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Huan Wang
- Department of Health Management Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Rui Ran
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yicheng Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yang Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
50
|
Pritchett EM, Van Goor A, Schneider BK, Young M, Lamont SJ, Schmidt CJ. Chicken pituitary transcriptomic responses to acute heat stress. Mol Biol Rep 2023; 50:5233-5246. [PMID: 37127810 DOI: 10.1007/s11033-023-08464-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Poultry production is vulnerable to increasing temperatures in terms of animal welfare and in economic losses. With the predicted increase in global temperature and the number and severity of heat waves, it is important to understand how chickens raised for food respond to heat stress. This knowledge can be used to determine how to select chickens that are adapted to thermal challenge. As neuroendocrine organs, the hypothalamus and pituitary provide systemic regulation of the heat stress response. METHODS AND RESULTS Here we report a transcriptome analysis of the pituitary response to acute heat stress. Chickens were stressed for 2 h at 35 °C (HS) and transcriptomes compared with birds maintained in thermoneutral temperatures (25 °C). CONCLUSIONS The observations were evaluated in the context of ontology terms and pathways to describe the pituitary response to heat stress. The pituitaries of heat stressed birds exhibited responses to hyperthermia through altered expression of genes coding for chaperones, cell cycle regulators, cholesterol synthesis, transcription factors, along with the secreted peptide hormones, prolactin, and proopiomelanocortin.
Collapse
Affiliation(s)
| | - Angelica Van Goor
- Animal Science, Iowa State University, Ames, IA, USA
- Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | | | - Meaghan Young
- Animal and Food Science, University of Delaware, Newark, DE, USA
| | | | - Carl J Schmidt
- Animal and Food Science, University of Delaware, Newark, DE, USA.
| |
Collapse
|