1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Saqirile, Deng Y, Li K, Yan W, Li K, Wang C. Gene Expression Regulation and the Signal Transduction of Programmed Cell Death. Curr Issues Mol Biol 2024; 46:10264-10298. [PMID: 39329964 DOI: 10.3390/cimb46090612] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Cell death is of great significance in maintaining tissue homeostasis and bodily functions. With considerable research coming to the fore, it has been found that programmed cell death presents in multiple modalities in the body, which is not only limited to apoptosis, but also can be divided into autophagy, pyroptosis, ferroptosis, mitotic catastrophe, entosis, netosis, and other ways. Different forms of programmed cell death have disparate or analogous characteristics with each other, and their occurrence is accompanied by multiple signal transduction and the role of a myriad of regulatory factors. In recent years, scholars across the world have carried out considerable in-depth research on programmed cell death, and new forms of cell death are being discovered continually. Concomitantly, the mechanisms of intricate signaling pathways and regulators have been discovered. More critically, cancer cells tend to choose distinct ways to evade cell death, and different tumors adapt to different manners of death. Therefore, targeting the cell death network has been regarded as an effective tumor treatment strategy for a long time. The objective of our paper is to review the signaling pathways and gene regulation in several typical types of programmed cell death and their correlation with cancer.
Collapse
Affiliation(s)
- Saqirile
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Yuxin Deng
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Kexin Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Wenxin Yan
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Ke Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| |
Collapse
|
3
|
Huang K, Yu L, Lu D, Zhu Z, Shu M, Ma Z. Long non-coding RNAs in ferroptosis, pyroptosis and necroptosis: from functions to clinical implications in cancer therapy. Front Oncol 2024; 14:1437698. [PMID: 39267831 PMCID: PMC11390357 DOI: 10.3389/fonc.2024.1437698] [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: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
As global population ageing accelerates, cancer emerges as a predominant cause of mortality. Long non-coding RNAs (lncRNAs) play crucial roles in cancer cell growth and death, given their involvement in regulating downstream gene expression levels and numerous cellular processes. Cell death, especially non-apoptotic regulated cell death (RCD), such as ferroptosis, pyroptosis and necroptosis, significantly impacts cancer proliferation, invasion and metastasis. Understanding the interplay between lncRNAs and the diverse forms of cell death in cancer is imperative. Modulating lncRNA expression can regulate cancer onset and progression, offering promising therapeutic avenues. This review discusses the mechanisms by which lncRNAs modulate non-apoptotic RCDs in cancer, highlighting their potential as biomarkers for various cancer types. Elucidating the role of lncRNAs in cell death pathways provides valuable insights for personalised cancer interventions.
Collapse
Affiliation(s)
- Ke Huang
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Li Yu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Dingci Lu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Ziyi Zhu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Min Shu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Zhaowu Ma
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
4
|
Jasielski P, Zawlik I, Bogaczyk A, Potocka N, Paszek S, Maźniak M, Witkoś A, Korzystka A, Kmieć A, Kluz T. The Promotive and Inhibitory Role of Long Non-Coding RNAs in Endometrial Cancer Course-A Review. Cancers (Basel) 2024; 16:2125. [PMID: 38893244 PMCID: PMC11171405 DOI: 10.3390/cancers16112125] [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: 04/29/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Endometrial cancer is one of the most common malignant tumours in women. The development of this tumour is associated with several genetic disorders, many of which are still unknown. One type of RNA molecules currently being intensively studied in many types of cancer are long non-coding RNAs (lncRNAs). LncRNA-coding genes occupy a large fraction of the human genome. LncRNAs regulate many aspects of cell development, metabolism, and other physiological processes. Diverse types of lncRNA can function as a tumour suppressor or an oncogene that can alter migration, invasion, cell proliferation, apoptosis, and immune system response. Recent studies suggest that selected lncRNAs are important in an endometrial cancer course. Our article describes over 70 lncRNAs involved in the development of endometrial cancer, which were studied via in vivo and in vitro research. It was proved that lncRNAs could both promote and inhibit the development of endometrial cancer. In the future, lncRNAs may become an important therapeutic target. The aim of this study is to review the role of lncRNAs in the development of carcinoma of uterine body.
Collapse
Affiliation(s)
- Patryk Jasielski
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Izabela Zawlik
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Anna Bogaczyk
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Natalia Potocka
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Sylwia Paszek
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Michał Maźniak
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Aleksandra Witkoś
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Adrianna Korzystka
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Aleksandra Kmieć
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
| | - Tomasz Kluz
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
- Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| |
Collapse
|
5
|
Cong P, Shang B, Zhang L, Wu Z, Wang Y, Li J, Zhang L. New insights into the treatment of polycystic ovary syndrome: HKDC1 promotes the growth of ovarian granulocyte cells by regulating mitochondrial function and glycolysis. J Mol Histol 2024; 55:187-199. [PMID: 38478190 DOI: 10.1007/s10735-024-10183-8] [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: 06/01/2023] [Accepted: 02/06/2024] [Indexed: 04/05/2024]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disease, and its pathogenesis and treatment are still unclear. Hexokinase domain component 1 (HKDC1) participates in regulating mitochondrial function and glycolysis. However, its role in PCOS development remains unrevealed. Here, female C57BL/6 mice were intraperitoneally injected with dehydroepiandrosterone (DHEA; 60 mg/kg body weight) to establish an in vivo model of PCOS. In vitro, KGN cells, a human ovarian granular cell line, were used to explore the potential mechanisms. DHEA-treated mice exhibited a disrupted estrus cycle, abnormal hormone levels, and insulin resistance. Dysfunction in mitochondria and glycolysis is the main reason for PCOS-related growth inhibition of ovarian granular cells. Here, we found that the structure of mitochondria was impaired, less ATP was generated and more mitochondrial Reactive Oxygen Species were produced in HKDC1-silenced KGN cells. Moreover, HKDC1 knockdown inhibited glucose consumption and decreased the production of glucose-6-phosphate and lactic acid. Conclusively, HKDC1 protects ovarian granulocyte cells from DHEA-related damage at least partly by preserving mitochondrial function and maintaining glycolysis.
Collapse
Affiliation(s)
- Peiwei Cong
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Bing Shang
- Chinese Medicine Literature Research Institute, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Lina Zhang
- Teaching and Experiment Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Zhaoli Wu
- College of Acupuncture and Massage, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yanan Wang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jia Li
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Lin Zhang
- College of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China.
| |
Collapse
|
6
|
Yin C, Li J, Li S, Yang X, Lu Y, Wang C, Liu B. LncRNA-HOXC-AS2 regulates tumor-associated macrophage polarization through the STAT1/SOCS1 and STAT1/CIITA pathways to promote the progression of non-small cell lung cancer. Cell Signal 2024; 115:111031. [PMID: 38168631 DOI: 10.1016/j.cellsig.2023.111031] [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: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Tumor-associated macrophages (TAMs) mainly exhibit the characteristics of M2-type macrophages, and the regulation of TAM polarization is a new target for cancer therapy, among which lncRNAs are key regulatory molecules. This study aimed to explore the effects of lncRNA-HOXC-AS2 on non-small cell lung cancer (NSCLC) by regulating TAM polarization. THP-1 cells were used to differentiate into macrophages, and TAMs were obtained by coculture with A549 cells. The M1/M2 cell phenotype and HOXC-AS2 expression were detected, and A549-derived exosomes (A549-exo) were used to elucidate the effects of A549 on macrophage polarization and HOXC-AS2 expression. Then, by interfering with HOXC-AS2 or STAT1, the effects of HOXC-AS2 regulation of STAT1 on the TAM phenotype and STAT1/SOCS1 and STAT1/CIITA pathways were analyzed, and the proliferation and metastasis of NSCLC cells in the coculture system were also detected. Results showed that HOXC-AS2 expression in M2 macrophages and TAMs was significantly higher than that in M1 macrophages, and A549-exo promoted HOXC-AS2 expression and M2 polarization. Intervention HOXC-AS2 resulted in increased M1 marker expression, decreased M2 marker expression, and activation of STAT1/SOCS1 and STAT1/CIITA pathways in TAMs. In addition, HOXC-AS2 was mainly expressed in the cytoplasm of TAMs and could bind to STAT1. Further experiments confirmed that intervention HOXC-AS2 promoted the M1 polarization of TAMs by targeting STAT1 and weakened the promoting effects of TAMs on the proliferation and metastasis of NSCLC. In conclusion, HOXC-AS2 inhibited the activation of STAT1/SOCS1 and STAT1/CIITA pathways and promoted M2 polarization of TAMs by binding with STAT1, thus promoting NSCLC.
Collapse
Affiliation(s)
- Cunli Yin
- School of Medicine, University of Electronic Science and Technology of China, China
| | - Jing Li
- Department of General Internal Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China Chengdu, China
| | - Siru Li
- School of Medicine, University of Electronic Science and Technology of China, China
| | - Xi Yang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, China
| | - Yingchun Lu
- School of Medicine, University of Electronic Science and Technology of China, China
| | - Chunyu Wang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, China
| | - Bin Liu
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China Chengdu, China.
| |
Collapse
|
7
|
Chen HY, Li XN, Yang L, Ye CX, Chen ZL, Wang ZJ. CircVMP1 promotes glycolysis and disease progression by upregulating HKDC1 in colorectal cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:1617-1630. [PMID: 38009649 DOI: 10.1002/tox.24061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to play important roles in cancers. Here, we characterized circVMP1 (hsa_circ_0006508), an important circRNA which promoted glycolysis and disease progression in colorectal cancer (CRC). In this study, we aimed to explore the mechanism by which circVMP1 regulated tumor glycolysis and its related pathways in promoting CRC cell proliferation and metastasis. METHODS The expression level of circVMP1 in CRC tissues and adjacent normal tissues was detected using quantitative PCR. In vitro and in vivo functional experiments were used to evaluate the effects of circVMP1 in the regulation of CRC cell proliferation and migration. Mitochondrial stress tests and glycolysis stress tests were conducted to detect the effect of circVMP1 on oxidative phosphorylation and glycolysis. Dual-luciferase reporter and RNA immunoprecipitation assays were used to evaluate the interaction between circVMP1, miR-3167, and HKDC1. RESULTS We demonstrated that the level of circVMP1 was significantly upregulated in CRC tissues compared with normal tissues. In HCT116 and SW480 cells, overexpression of circVMP1 promoted proliferation, metastasis, and glycolysis. In vivo analysis indicated that circVMP1 accelerated the proliferation of xenograft tumors. As for the mechanism, overexpression of circVMP1 increased the levels of hexokinase domain component 1 (HKDC1) through competitive binding with miR-3167. CONCLUSION Our study reported that circVMP1 was one of the tumor driver genes that promoted CRC malignant progression and glycolysis by upregulating HKDC1. CircVMP1/miR-3167/HKDC1 was a signaling axis that might be a target for CRC therapy.
Collapse
Affiliation(s)
- Hong-Yu Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiang-Nan Li
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chun-Xiang Ye
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhi-Lei Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhen-Jun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Xiang Y, Huang G, Wang J, Hua Q. lncRNA HOXC-AS2 promotes the progression of hypopharyngeal cancer by binding to the P62 protein mediating the autophagy process. Aging (Albany NY) 2023; 15:12476-12496. [PMID: 37944249 PMCID: PMC10683610 DOI: 10.18632/aging.205192] [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: 07/29/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
Hypopharyngeal carcinoma is the most malignant type of head and neck squamous cell carcinoma, and lncRNAs play an important role in its formation and progression. However, the related specific mechanisms are rarely studied. lncRNAs closely associated with hypopharyngeal cancer were examined by lncRNA sequencing for in-depth exploration of the relationship between HOXC-AS2 and hypopharyngeal cancer pathogenesis. The mRNA expression of HOXC-AS2 and related genes was measured by qRT-PCR, and the biological function of HOXC-AS2 in hypopharyngeal carcinoma was demonstrated by gain- and loss-of-function experiments. RNA pulldown, RNA immunoprecipitation (RIP) and gene body truncation experiments and transcriptome sequencing were used to investigate the potential mechanism of HOXC-AS2 and its downstream genes, including P62, NF-KB and HMOX1. Finally, the biological function of HOXC-AS2 was confirmed in animal experiments. HOXC-AS2 and P62 expression was significantly upregulated in hypopharyngeal cancer tissues compared with normal hypopharyngeal tissues, while HMOX1 expression was decreased. Functionally, HOXC-AS2 overexpression can promote the viability, proliferation, migration and invasion of hypopharyngeal cancer cells and facilitate hypopharyngeal cancer progression. It was confirmed that HOXC-AS2 can bind to the P62 protein and activate the NF-KB signaling pathway, thereby affecting HMOX1 expression and regulating autophagy in hypopharyngeal cancer cells, ultimately regulating the formation and progression of hypopharyngeal cancer. In conclusion, our findings suggest that HOXC-AS2 regulates the progression of hypopharyngeal cancer by regulating autophagy and is abnormally highly expressed in hypopharyngeal cancer tissues. HOXC-AS2 may become a new target for the diagnosis and treatment of hypopharyngeal cancer.
Collapse
Affiliation(s)
- Yuandi Xiang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Guoquan Huang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
- Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, Hubei 445000, China
- Department of Gastrointestinal Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
9
|
Peng T, Zhang C, Chen WJ, Zhao XF, Wu WB, Yang WJ, Liang RJ. Pyroptosis: the dawn of a new era in endometrial cancer treatment. Front Oncol 2023; 13:1277639. [PMID: 37965452 PMCID: PMC10642841 DOI: 10.3389/fonc.2023.1277639] [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: 08/15/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Endometrial cancer (EC) is a malignancy of the inner epithelial lining of the uterus. While early-stage EC is often curable through surgery, the management of advanced, recurrent and metastatic EC poses significant challenges and is associated with a poor prognosis. Pyroptosis, an emerging form of programmed cell death, is characterized by the cleavage of gasdermin proteins, inducing the formation of extensive gasdermin pores in the cell membrane and the leakage of interleukin-1β (IL-1β) and interleukin-18 (IL-18), consequently causing cell swelling, lysis and death. It has been found to be implicated in the occurrence and progression of almost all tumors. Recent studies have demonstrated that regulating tumor cells pyroptosis can exploit synergies function with traditional tumor treatments. This paper provides an overview of the research progress made in molecular mechanisms of pyroptosis. It then discusses the role of pyroptosis and its components in initiation and progression of endometrial cancer, emphasizing recent insights into the underlying mechanisms and highlighting unresolved questions. Furthermore, it explores the potential value of pyroptosis in the treatment of endometrial cancer, considering its current application in tumor radiotherapy, chemotherapy, targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Tian Peng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wen-Jun Chen
- School of Nursing, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Gynaecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xue-Fei Zhao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Bo Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Ji Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruo-Jia Liang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gynaecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Zhang RN, Jing ZQ, Zhang L, Sun ZJ. Epigenetic regulation of pyroptosis in cancer: Molecular pathogenesis and targeting strategies. Cancer Lett 2023; 575:216413. [PMID: 37769798 DOI: 10.1016/j.canlet.2023.216413] [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: 07/19/2023] [Revised: 09/02/2023] [Accepted: 09/23/2023] [Indexed: 10/02/2023]
Abstract
Immune checkpoint blockade therapy has revolutionized the field of cancer treatment, leading to durable responses in patients with advanced and metastatic cancers where conventional therapies were insufficient. However, factors like immunosuppressive cells and immune checkpoint molecules within the tumor microenvironment (TME) can suppress the immune system and thus negatively affect the efficiency of immune checkpoint inhibitors. Pyroptosis, a gasdermin-induced programmed cell death, could transform "cold tumors" to "hot tumors" to improve the milieu of TME, thus enhancing the immune response and preventing tumor growth. Recently, evidence showed that epigenetics could regulate pyroptosis, which further affects tumorigenesis, suggesting that epigenetics-based tumor cells pyroptosis could be a promising therapeutic strategy. Hence, this review focuses on the pyroptotic mechanism and summarizes three common types of epigenetics, DNA methylation, histone modification, and non-coding RNA, all of which have a role in regulating the expression of transcription factors and proteins involved in pyroptosis in cancer. Especially, we discuss targeting strategies on epigenetic-regulated pyroptosis and provide insights on the future trend of cancer research which may fuel cancer therapies into a new step.
Collapse
Affiliation(s)
- Ruo-Nan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, China
| | - Zhi-Qian Jing
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, China; Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Wang J, Hua S, Bao H, Yuan J, Zhao Y, Chen S. Pyroptosis and inflammasomes in cancer and inflammation. MedComm (Beijing) 2023; 4:e374. [PMID: 37752941 PMCID: PMC10518439 DOI: 10.1002/mco2.374] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Nonprogrammed cell death (NPCD) and programmed cell death (PCD) are two types of cell death. Cell death is significantly linked to tumor development, medication resistance, cancer recurrence, and metastatic dissemination. Therefore, a comprehensive understanding of cell death is essential for the treatment of cancer. Pyroptosis is a kind of PCD distinct from autophagy and apoptosis in terms of the structure and function of cells. The defining features of pyroptosis include the release of an inflammatory cascade reaction and the expulsion of lysosomes, inflammatory mediators, and other cellular substances from within the cell. Additionally, it displays variations in osmotic pressure both within and outside the cell. Pyroptosis, as evidenced by a growing body of research, is critical for controlling the development of inflammatory diseases and cancer. In this paper, we reviewed the current level of knowledge on the mechanism of pyroptosis and inflammasomes and their connection to cancer and inflammatory diseases. This article presents a theoretical framework for investigating the potential of therapeutic targets in cancer and inflammatory diseases, overcoming medication resistance, establishing nanomedicines associated with pyroptosis, and developing risk prediction models in refractory cancer. Given the link between pyroptosis and the emergence of cancer and inflammatory diseases, pyroptosis-targeted treatments may be a cutting-edge treatment strategy.
Collapse
Affiliation(s)
- Jie‐Lin Wang
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Sheng‐Ni Hua
- Department of Radiation OncologyZhuhai Peoples HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Hai‐Juan Bao
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jing Yuan
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yang Zhao
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Shuo Chen
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
12
|
Zhao P, Yuan F, Xu L, Jin Z, Liu Y, Su J, Yuan L, Peng L, Wang C, Zhang G. HKDC1 reprograms lipid metabolism to enhance gastric cancer metastasis and cisplatin resistance via forming a ribonucleoprotein complex. Cancer Lett 2023:216305. [PMID: 37423558 DOI: 10.1016/j.canlet.2023.216305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/21/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
As essential modulators of transcription and translation, RNA-binding proteins (RBPs) are frequently dysregulated in cancer. Bioinformatics study reveals that the RNA-binding protein hexokinase domain component 1 (HKDC1) is overexpressed in gastric cancer (GC). As HKDC1 plays a role in lipid homeostasis in the liver and glucose metabolism in certain cancers, the exact mechanism of action of HKDC1 in GC remains largely unknown. Upregulation of HKDC1 correlates with chemoresistance and poor prognosis in GC patients. HKDC1 enhances invasion, migration and resistance to cisplatin (CDDP) in GC cells in vitro and in vivo. Comprehensive transcriptomic sequencing and metabolomic analysis reveal that HKDC1 mediates abnormal lipid metabolism in GC cells. Herein, we identify a number of HKDC1-binding endogenous RNAs in GC cells, including protein kinase, DNA-activated, catalytic subunit (PRKDC) mRNA. We further validate that PRKDC is a crucial downstream effector of HKDC1 induced-GC tumorigenesis depends on lipid metabolism. Interestingly, G3BP1, a well-known oncoprotein, can be bound by HKDC1. HKDC1 cooperates with G3BP1 to enhance the stability of PRKDC transcript. Our results reveal a novel HKDC1/G3BP1-PRKDC regulatory axis that induces GC metastasis and chemoresistance via reprogramming lipid metabolism, which may provide an effective therapeutic strategy for a subset of GC with HKDC1 overexpression.
Collapse
Affiliation(s)
- Ping Zhao
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Lijuan Xu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhenghao Jin
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yang Liu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Jing Su
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, 221009, China
| | - Lin Yuan
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lei Peng
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China.
| | - Guoxin Zhang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
13
|
Bhat SA, Farooq Z, Ismail H, Corona-Avila I, Khan MW. Unraveling the Sweet Secrets of HCC: Glucometabolic Rewiring in Hepatocellular Carcinoma. Technol Cancer Res Treat 2023; 22:15330338231219434. [PMID: 38083797 PMCID: PMC10718058 DOI: 10.1177/15330338231219434] [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: 09/20/2023] [Revised: 11/06/2023] [Accepted: 11/13/2017] [Indexed: 12/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the primary form of liver cancer. It causes ∼ 800 000 deaths per year, which is expected to increase due to increasing rates of obesity and metabolic dysfunction associated steatotic liver disease (MASLD). Current therapies include immune checkpoint inhibitors, tyrosine kinase inhibitors, and monoclonal antibodies, but these therapies are not satisfactorily effective and often come with multiple side effects and recurrences. Metabolic reprogramming plays a significant role in HCC progression and is often conserved between tumor types. Thus, targeting rewired metabolic pathways could provide an attractive option for targeting tumor cells alone or in conjunction with existing treatments. Therefore, there is an urgent need to identify novel targets involved in cancer-mediated metabolic reprogramming in HCC. In this review, we provide an overview of molecular rewiring and metabolic reprogramming of glucose metabolism in HCC to understand better the concepts that might widen the therapeutic window against this deadly cancer.
Collapse
Affiliation(s)
- Sheraz Ahmad Bhat
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
- Sri Pratap College, Cluster University Srinagar, Srinagar, Jammu & Kashmir, India
| | - Zeenat Farooq
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Hagar Ismail
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Irene Corona-Avila
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Md. Wasim Khan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Identification of m7G Methylation-Related miRNA Signature Associated with Survival and Immune Microenvironment Regulation in Uterine Corpus Endometrial Carcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8776678. [DOI: 10.1155/2022/8776678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 11/27/2022]
Abstract
Background. N7-methylguanosine (m7G) has been implicated in the development of cancer. The role of m7G-related miRNAs in the survival prediction of UCEC patients has not been investigated. Current research was the first to construct an m7G-related miRNA model to accurately predict the survival of patients with uterine corpus endometrial carcinoma (UCEC) and to explore immune cell infiltration and immune activity in the tumor microenvironment. Methods. RNA-seq data and clinical information of UCEC patients were derived from The Cancer Genome Atlas (TCGA) database. Using the TargetScan online database, we predicted miRNAs linked to the m7G-related genes and identified miRNAs which were significantly associated with the survival in UCEC patients and constructed a risk scoring model. The TCGA-UCEC cases were scored according to the risk model, and the high- and low-risk groups were divided by the median risk value. Gene enrichment analysis and immune cell infiltration and immune function analysis were performed using “clusterProfiler” and “GSVA” packages in R. Results. The survival prediction model consisted of 9 miRNAs, namely, hsa-miR-1301, hsa-miR-940, hsa-miR-592, hsa-miR-3170, hsa-miR-876, hsa-miR-215, hsa-miR-934, hsa-miR-3920, and hsa-miR-216b. Survival of UCEC patients in the high-risk group was worse than that in the low-risk group (
). The receiver operating characteristic (ROC) curve showed that the model had good predictive performance, and the area under the curve was 0.800, 0.690, and 0.705 for 1-, 3-, and 5-year survival predictions, respectively. There were differences in the degree of immune cell infiltration and immune activity between the low-risk and high-risk groups. The expression levels of the identified differentially expressed genes correlated with the susceptibility to multiple anticancer drugs. Conclusions. The survival prediction model constructed based on 9 m7G-related miRNAs had good predictive performance.
Collapse
|
15
|
Cheng Y, Wu X, Xia Y, Liu W, Wang P. The role of lncRNAs in regulation of DKD and diabetes-related cancer. Front Oncol 2022; 12:1035487. [PMID: 36313695 PMCID: PMC9606714 DOI: 10.3389/fonc.2022.1035487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetes mellitus often results in several complications, such as diabetic kidney disease (DKD) and end-stage renal diseases (ESRDs). Cancer patients often have the dysregulated glucose metabolism. Abnormal glucose metabolism can enhance the tumor malignant progression. Recently, lncRNAs have been reported to regulate the key proteins and signaling pathways in DKD development and progression and in cancer patients with diabetes. In this review article, we elaborate the evidence to support the function of lncRNAs in development of DKD and diabetes-associated cancer. Moreover, we envisage that lncRNAs could be diagnosis and prognosis biomarkers for DKD and cancer patients with diabetes. Furthermore, we delineated that targeting lncRNAs might be an alternative approach for treating DKD and cancer with dysregulated glucose metabolism.
Collapse
Affiliation(s)
- Yawei Cheng
- Department of Disease Prevention, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
- *Correspondence: Yawei Cheng, ; Peter Wang,
| | - Xiaowen Wu
- Department of Disease Prevention, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, China
| | - Yujie Xia
- Department of Food Science and Technology Centers, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Wenjun Liu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
- *Correspondence: Yawei Cheng, ; Peter Wang,
| |
Collapse
|