1
|
Haag JG, Shen X, Sinha N, Chen S, Deng B, Zhang H, John C, Sun W, Emanuele M, Zhou C, Bae-Jump V. Onvansertib inhibits cell proliferation and increases sensitivity to paclitaxel in uterine serous cancer cells. Am J Cancer Res 2025; 15:1719-1732. [PMID: 40371133 PMCID: PMC12070104 DOI: 10.62347/lizg3616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/08/2025] [Indexed: 05/16/2025] Open
Abstract
Uterine serous carcinoma (USC) belongs to the non-endometrioid subtype of endometrial cancer that is known for its highly aggressive behavior and poor prognosis, highlighting the warrant of novel strategies for the treatment of USC. PLK1 is a type of serine/threonine kinase that is crucial for controlling the progression of the cell cycle, DNA damage response, and genome stability. Targeting PLK1 exhibits potent anti-tumorigenic activity in pre-clinical models of multiple cancer types, and several PLK1 inhibitors have shown significant clinical benefit and favorable safety profiles alone or in combination with other chemotherapeutic agents. Onvansertib is an oral, selective PLK1 inhibitor that exhibits anti-proliferative activity in multiple types of cancer cell and animal models and has demonstrated clinical activity and a favorable safety profile in recent clinical trials. Hence, we investigated the anti-tumorigenic effects of onvansertib in USC cell lines. Nanomolar concentrations of onvansertib significantly inhibited cellular proliferation, led to cell cycle G2 arrest, induced cellular stress and apoptosis, caused DNA damage, and reduced cell adhesion and invasion in ARK-1 and SPEC-2 cells. The combination of onvansertib with paclitaxel demonstrated a synergistic effect in cell proliferation inhibition via inducing cell apoptosis and DNA damage. Our results provide preclinical evidence that onvansertib may be an effective strategy to treat USC and deserves further evaluation in animal models and clinical trials.
Collapse
Affiliation(s)
- Jennifer G Haag
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Xiaochang Shen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care HospitalBeijing 100026, P. R. China
| | - Nikita Sinha
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Shuning Chen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care HospitalBeijing 100026, P. R. China
| | - Boer Deng
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care HospitalBeijing 100026, P. R. China
| | - Haomeng Zhang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care HospitalBeijing 100026, P. R. China
| | - Catherine John
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Michael Emanuele
- Department of Pharmacology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Liang H, Fang C, Qiu M. The multi-target mechanism of action of Selaginella doederleinii Hieron in the treatment of nasopharyngeal carcinoma: a network pharmacology and multi-omics analysis. Sci Rep 2025; 15:159. [PMID: 39747499 PMCID: PMC11695685 DOI: 10.1038/s41598-024-83921-3] [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: 08/20/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) presents significant treatment challenges due to its complex etiology and late-stage diagnosis. The traditional Chinese medicine Selaginella doederleinii Hieron (S. doederleinii) has shown potentiality in NPC treatment due to its multi-target, multi-pathway anti-cancer mechanisms. First, we identified NPC related target genes from databases like GeneCards, OMIM, and DisGeNET, and performed WGCNA analysis on the GSE53819 dataset to identify several important gene modules related to NPC. Active components and their targets in S. doederleinii were screened from the TCMSP and other databases, identifying 32 overlapping genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that these genes are primarily involved in critical biological processes like protein phosphorylation and cell cycle regulation. A protein-protein interaction network was constructed, and cytoHubba identified six key genes (BCL2, MAPK14, ABCB1, PLK1, ATM, HMOX1). Kaplan-Meier analysis and immune infiltration analysis further showed that these key genes are closely related to the prognosis and immune microenvironment of NPC patients. Single-cell RNA sequencing analysis revealed the expression distribution of key genes across different immune cell types and explored their roles in the differentiation process of malignant cells through pseudotime trajectory analysis. Molecular docking and dynamics simulation results indicated that the Berberine-MAPK14 and Matairesinol-PLK1 complexes have high binding affinity and stability. Binding free energy calculations confirmed the stability of these complexes. Based on our comprehensive multi-level analysis, the active components of S. doederleinii may play a significant role in the treatment of NPC through multi-pathway and multi-target synergistic effects.
Collapse
Affiliation(s)
- Huaguo Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Caifu Fang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Meng Qiu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
3
|
Zhou YY, Zhao SY, Huang FJ, Zhang LJ, Liu YL, Wang J, Ma XJ. JPT2 in subclinical hypothyroidism-related miscarriage as a transcription co-factor: involvement of LEPR/STAT3 activation. J Endocrinol Invest 2024; 47:2521-2537. [PMID: 38907823 DOI: 10.1007/s40618-024-02343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/18/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND AND PURPOSE Subclinical hypothyroidism (SCH) has been identified to be associated with implantation failure, in which the dysfunction of trophoblast cells is involved. In this study, the transcriptomics of aborted placenta from SCH rats were analyzed. Jupiter microtubule-associated homolog 2 (JPT2) was downregulated in the aborted placenta. This study aims to investigate its role in SCH-associated miscarriage. METHODS Spontaneous abortion was observed in SCH rats generated by thyroidectomy combined with levothyroxine administration. The transcriptomics analysis was performed using aborted placenta. Afterward, the effects of JPT2 on trophoblast cells were explored using gain-and loss-of-function experiments. RESULTS Transcriptomics analysis showed 1286 downregulated genes and 2300 upregulated genes in the aborted placenta, and JPT2 was significantly downregulated in the aborted placenta from SCH rats. Afterward, gain-and loss-of-function experiments exhibited that overexpression of JPT2 promoted the proliferation, migration, invasion, spheroid formation of HTR-8/SVneo trophoblast cells and their attachment to endometrial stromal cells, while these biological behaviors were suppressed by JPT2 knockdown. Furthermore, JPT2 accelerated the transcription of leptin receptor (LEPR), and activated signal transducer and activator of transcription 3 (STAT3) signal in a transcription factor AP-2γ-dependent manner. In addition, silencing of LEPR abolished the role of JPT2. CONCLUSION Our results revealed that JPT2, which was downregulated in the aborted placenta from SCH rats, promoted proliferation, migration, invasion, spheroid formation, and attachment of trophoblast cells via regulating LEPR/STAT3 axis as a transcription co-factor. It is indicated that low expression of JPT2 may contribute to the abortion in individuals with SCH.
Collapse
Affiliation(s)
- Y-Y Zhou
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - S-Y Zhao
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - F-J Huang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - L-J Zhang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - Y-L Liu
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - J Wang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - X-J Ma
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China.
| |
Collapse
|
4
|
Yun F, Wu N, Yi X, Zhang X, Feng Y, Ni Q, Gai Y, Li E, Yang Z, Zhang Q, Sai B, Kuang Y, Zhu Y. NOD2 reduces the chemoresistance of melanoma by inhibiting the TYMS/PLK1 signaling axis. Cell Death Dis 2024; 15:720. [PMID: 39353904 PMCID: PMC11445241 DOI: 10.1038/s41419-024-07104-8] [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: 04/25/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
Nucleotide-binding oligomerization domain 2 (NOD2) is an immune sensor crucial for eliciting the innate immune responses. Nevertheless, discrepancies exist regarding the effect of NOD2 on different types of cancer. This study aimed to investigate these function of NOD2 in melanoma and its underlying mechanisms. We have validated the tumor suppressor effect of NOD2 in melanoma. NOD2 inhibited the proliferation of melanoma cells, hindering their migration and invasion while promoting the onset of apoptosis. Our study showed that NOD2 expression is closely related to pyrimidine and folate metabolism. NOD2 inhibits thymidylate synthase (TYMS) expression by promoting K48-type ubiquitination modification of TYMS, thereby decreasing the resistance of melanoma cells to 5-fluorouracil (5-FU) and capecitabine (CAP). TYMS was identified to form a complex with Polo-like Kinase 1 (PLK1) and activate the PLK1 signaling pathway. Furthermore, we revealed that the combination of the PLK1 inhibitor volasertib (BI6727) with 5-FU or CAP had a synergistic effect repressing the proliferation, migration, and autophagy of melanoma cells. Overall, our research highlights the protective role of NOD2 in melanoma and suggests that targeting NOD2 and the TYMS/PLK1 signaling axis is a high-profile therapy that could be a prospect for melanoma treatment.
Collapse
Affiliation(s)
- Fang Yun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Na Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xiaojia Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuedan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yu Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Qinxuan Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yanlong Gai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Enjiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Zhe Yang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Buqing Sai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yingmin Kuang
- Department of Organ Transplantation, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Khan IR, Sadida HQ, Hashem S, Singh M, Macha MA, Al-Shabeeb Akil AS, Khurshid I, Bhat AA. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed Pharmacother 2024; 176:116873. [PMID: 38843587 DOI: 10.1016/j.biopha.2024.116873] [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: 03/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Esophageal cancer (EC) is significantly influenced by the tumor microenvironment (TME) and altered signaling pathways. Downregulating these pathways in EC is essential for suppressing tumor development, preventing metastasis, and enhancing therapeutic outcomes. This approach can increase tumor sensitivity to treatments, enhance patient outcomes, and inhibit cancer cell proliferation and spread. The TME, comprising cellular and non-cellular elements surrounding the tumor, significantly influences EC's development, course, and treatment responsiveness. Understanding the complex relationships within the TME is crucial for developing successful EC treatments. Immunotherapy is a vital TME treatment for EC. However, the heterogeneity within the TME limits the application of anticancer drugs outside clinical settings. Therefore, identifying reliable microenvironmental biomarkers that can detect therapeutic responses before initiating therapy is crucial. Combining approaches focusing on EC signaling pathways with TME can enhance treatment outcomes. This integrated strategy aims to interfere with essential signaling pathways promoting cancer spread while disrupting factors encouraging tumor development. Unraveling aberrant signaling pathways and TME components can lead to more focused and efficient treatment approaches, identifying specific cellular targets for treatments. Targeting the TME and signaling pathways may reduce metastasis risk by interfering with mechanisms facilitating cancer cell invasion and dissemination. In conclusion, this integrative strategy has significant potential for improving patient outcomes and advancing EC research and therapy. This review discusses the altered signaling pathways and TME in EC, focusing on potential future therapeutics.
Collapse
Affiliation(s)
- Inamu Rashid Khan
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine Doha 26999, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir 192122, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Ibraq Khurshid
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar.
| |
Collapse
|
6
|
Chen X, Song QL, Ji R, Wang JY, Cao ML, Guo DY, Zhang Y, Yang J. JPT2 Affects Trophoblast Functions and Macrophage Polarization and Metabolism, and Acts as a Potential Therapeutic Target for Recurrent Spontaneous Abortion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306359. [PMID: 38417123 PMCID: PMC11040346 DOI: 10.1002/advs.202306359] [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: 09/04/2023] [Revised: 01/12/2024] [Indexed: 03/01/2024]
Abstract
Recurrent spontaneous abortion (RSA) is a pregnancy-related condition with complex etiology. Trophoblast dysfunction and abnormal macrophage polarization and metabolism are associated with RSA; however, the underlying mechanisms remain unknown. Jupiter microtubule-associated homolog 2 (JPT2) is essential for calcium mobilization; however, its role in RSA remains unclear. In this study, it is found that the expression levels of JPT2, a nicotinic acid adenine dinucleotide phosphate-binding protein, are decreased in the villous tissues of patients with RSA and placental tissues of miscarried mice. Mechanistically, it is unexpectedly found that abnormal JPT2 expression regulates trophoblast function and thus involvement in RSA via c-Jun N-terminal kinase (JNK) signaling, but not via calcium mobilization. Specifically, on the one hand, JPT2 deficiency inhibits trophoblast adhesion, migration, and invasion by inhibiting the JNK/atypical chemokine receptor 3 axis. On the other hand, trophoblast JPT2 deficiency contributes to M1 macrophage polarization by promoting the accumulation of citrate and reactive oxygen species via inhibition of the JNK/interleukin-6 axis. Self-complementary adeno-associated virus 9-JPT2 treatment alleviates embryonic resorption in abortion-prone mice. In summary, this study reveals that JPT2 mediates the remodeling of the immune microenvironment at the maternal-fetal interface, suggesting its potential as a therapeutic target for RSA.
Collapse
Affiliation(s)
- Xin Chen
- Reproductive Medical CenterRenmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic DevelopmentWuhanHubei430060China
| | - Qian Lin Song
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Rui Ji
- Reproductive Medical CenterRenmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic DevelopmentWuhanHubei430060China
| | - Jia Yu Wang
- Reproductive Medical CenterRenmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic DevelopmentWuhanHubei430060China
| | - Ming Liang Cao
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Duan Ying Guo
- Department of GynecologyLonggang District People's Hospital of ShenzhenShenzhen518172China
| | - Yan Zhang
- Department of Obstetrics and GynecologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Jing Yang
- Reproductive Medical CenterRenmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic DevelopmentWuhanHubei430060China
| |
Collapse
|
7
|
Xu R, Lee YJ, Kim CH, Min GH, Kim YB, Park JW, Kim DH, Kim JH, Yim H. Invasive FoxM1 phosphorylated by PLK1 induces the polarization of tumor-associated macrophages to promote immune escape and metastasis, amplified by IFITM1. J Exp Clin Cancer Res 2023; 42:302. [PMID: 37968723 PMCID: PMC10652615 DOI: 10.1186/s13046-023-02872-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Understanding the mechanism behind immune cell plasticity in cancer metastasis is crucial for identifying key regulators. Previously we found that mitotic factors regulate epithelial-mesenchymal transition, but how these factors convert to metastatic players in the tumor microenvironment (TME) is not fully understood. METHODS The clinical importance of mitotic factors was analyzed by heatmap analysis, a KM plot, and immunohistochemistry in lung adenocarcinoma (LUAD) patients. Immunoprecipitation, LC-MS/MS, kinase assay, and site-directed mutagenesis were performed for the interaction and phosphorylation. A tail-vein injection mouse model, Transwell-based 3D culture, microarray analysis, coculture with monocytes, and chromatin immunoprecipitation assays were used to elucidate the function of phosphorylated FoxM1 in metastasis of TME. RESULTS The phosphorylated FoxM1 at Ser25 by PLK1 acquires the reprogramming ability to stimulate the invasive traits in cancer and influence immune cell plasticity. This invasive form of p-FoxM1 upregulates the expression of IL1A/1B, VEGFA, and IL6 by direct activation, recruiting monocytes and promoting the polarization of M2d-like tumor-associated macrophages (TAMs). Upregulation of PD-L1 in LUAD having phosphomimetic FoxM1 facilitates immune evasion. In invasive LUAD with phosphomimetic FoxM1, IFITM1 is the most highly expressed through the activation of the STING-TBK1-IRF3 signaling, which enhances FoxM1-mediated signaling. Clinically, higher expression of FOXM1, PLK1, and IFITM1 is inversely correlated with the survival rate of advanced LUAD patients, providing a promising therapeutic strategy for the treatment of LUAD. CONCLUSION FoxM1-based therapy would be a potential therapeutic strategy for LUAD to reduce TAM polarization, immune escape, and metastasis, since FoxM1 functions as a genetic reprogramming factor reinforcing LUAD malignancy in the TME.
Collapse
Affiliation(s)
- Rong Xu
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Young-Joo Lee
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Chang-Hyeon Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Ga-Hong Min
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Yeo-Bin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Jung-Won Park
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Dae-Hoon Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Jung-Hyun Kim
- Division of Intractable Diseases Research, Department of Chronic Diseases Convergence Research, Korea National Institute of Health, Cheongju, Chungcheongbuk-Do, 28160, Republic of Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-Do, 15588, Republic of Korea.
| |
Collapse
|
8
|
Heßling LD, Troost-Kind B, Weiß M. NAADP-binding proteins - Linking NAADP signaling to cancer and immunity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119531. [PMID: 37394011 DOI: 10.1016/j.bbamcr.2023.119531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
NAADP is one of the most potent calcium mobilizing second messengers. Only recently, two NAADP-binding proteins have been identified: HN1L/JPT2 and LSM12. Further, ASPDH was suggested as a less selective binding partner. Apart from this newly uncovered link, little is known about the shared mechanisms between these proteins. The aim of this review is to assess potential functional connections between NAADP and its binding proteins. We here give a description of two major links. For one, HN1L/JPT2 and LSM12 both have potent oncogenic functions in several cancer types. Second, they are involved in similar cellular pathways in both cancer and immunity.
Collapse
Affiliation(s)
- Louisa D Heßling
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Berit Troost-Kind
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mariella Weiß
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
9
|
Wang Y, Yang W, Wang Q, Zhou Y. Mechanisms of esophageal cancer metastasis and treatment progress. Front Immunol 2023; 14:1206504. [PMID: 37359527 PMCID: PMC10285156 DOI: 10.3389/fimmu.2023.1206504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Esophageal cancer is a prevalent tumor of the digestive tract worldwide. The detection rate of early-stage esophageal cancer is very low, and most patients are diagnosed with metastasis. Metastasis of esophageal cancer mainly includes direct diffusion metastasis, hematogenous metastasis, and lymphatic metastasis. This article reviews the metabolic process of esophageal cancer metastasis and the mechanisms by which M2 macrophages, CAF, regulatory T cells, and their released cytokines, including chemokines, interleukins, and growth factors, form an immune barrier to the anti-tumor immune response mediated by CD8+ T cells, impeding their ability to kill tumor cells during tumor immune escape. The effect of Ferroptosis on the metastasis of esophageal cancer is briefly mentioned. Moreover, the paper also summarizes common drugs and research directions in chemotherapy, immunotherapy, and targeted therapy for advanced metastatic esophageal cancer. This review aims to serve as a foundation for further investigations into the mechanism and management of esophageal cancer metastasis.
Collapse
Affiliation(s)
- Yusheng Wang
- Department of Thoracic Surgery, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Wei Yang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Qianyun Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Yong Zhou
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
10
|
Jin C, Luo Y, Liang Z, Li X, Kołat D, Zhao L, Xiong W. Crucial role of the transcription factors family activator protein 2 in cancer: current clue and views. J Transl Med 2023; 21:371. [PMID: 37291585 PMCID: PMC10249218 DOI: 10.1186/s12967-023-04189-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
The transcription factor family activator protein 2 (TFAP2) is vital for regulating both embryonic and oncogenic development. The TFAP2 family consists of five DNA-binding proteins, including TFAP2A, TFAP2B, TFAP2C, TFAP2D and TFAP2E. The importance of TFAP2 in tumor biology is becoming more widely recognized. While TFAP2D is not well studied, here, we mainly focus on the other four TFAP2 members. As a transcription factor, TFAP2 regulates the downstream targets directly by binding to their regulatory region. In addition, the regulation of downstream targets by epigenetic modification, posttranslational regulation, and interaction with noncoding RNA have also been identified. According to the pathways in which the downstream targets are involved in, the regulatory effects of TFAP2 on tumorigenesis are generally summarized as follows: stemness and EMT, interaction between TFAP2 and tumor microenvironment, cell cycle and DNA damage repair, ER- and ERBB2-related signaling pathway, ferroptosis and therapeutic response. Moreover, the factors that affect TFAP2 expression in oncogenesis are also summarized. Here, we review and discuss the most recent studies on TFAP2 and its effects on carcinogenesis and regulatory mechanisms.
Collapse
Affiliation(s)
- Chen Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxiao Luo
- University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Chinese Academy for Medical Sciences Oxford Institute, Oxford, UK
| | - Xi Li
- Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Damian Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Linyong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Weixi Xiong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|