1
|
Long L, Tang X, Wang Y, Gu J, Xiong J, Luo H, Lv H, Zhou F, Cao K, Lin S. Network Pharmacology and Experimental Validation to Elucidate the Pharmacological Mechanisms of Luteolin Against Chondrocyte Senescence. Comb Chem High Throughput Screen 2025; 28:291-305. [PMID: 38299289 DOI: 10.2174/0113862073273675231114112804] [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: 07/23/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Luteolin, a flavonoid found in various medicinal plants, has shown promising antioxidant, anti-inflammatory, and anti-aging properties. The cartilaginous endplate (CEP) represents a crucial constituent of the intervertebral disc (IVD), assuming a pivotal responsibility in upholding both the structural and functional stability of the IVD. OBJECTIVE Exploring the precise mechanism underlying the protective effects of luteolin against senescence and degeneration of endplate chondrocytes (EPCs). METHODS Relevant targets associated with luteolin and aging were obtained from publicly available databases. To ascertain cellular functions and signaling pathways, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed. Core genes were identified through the construction of a protein-protein interaction (PPI) network. Molecular docking (MD) was utilized to assess the binding affinity of luteolin to these core genes. Finally, the impact of luteolin on the senescence and degeneration of EPCs was evaluated in an in vitro cellular senescence model induced by tert-butyl hydroperoxide (TBHP). RESULTS There are 145 overlapping targets between luteolin and senescence. Analysis using GO revealed that these targets primarily participate in cellular response to oxidative stress and reactive oxygen species. KEGG analysis demonstrated that these markers mainly associate with signaling pathways such as p53 and PI3K-Akt. MD simulations exhibited luteolin's binding affinity to P53, Cyclin-dependent kinase (CDK)2, and CDK4. Cell cycle, cell proliferation, and β- galactosidase assays confirmed that luteolin mitigated senescence in SW1353 cells. Western blot assays exhibited that luteolin significantly suppressed the expression of Matrix Metallopeptidase (MMP) 13, P53, and P21, while concurrently promoting CDK2, CDK4, and Collagen Type II Alpha 1 (COL2A1) expression. CONCLUSION In summary, luteolin demonstrated beneficial properties against aging and degeneration in EPCs, offering novel insights to mitigate the progression of intervertebral disc degeneration (IVDD).
Collapse
Affiliation(s)
- Ling Long
- Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Xiaokai Tang
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Yi Wang
- Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Jiaxiang Gu
- Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Jiachao Xiong
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Hao Luo
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Hao Lv
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Faxin Zhou
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Kai Cao
- The Orthopedics Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330209, China
| | - Sijian Lin
- The Rehabilitation Medicine Department, The Second Affiliated Hospital of Nanchang University, 330006, China
| |
Collapse
|
2
|
Mahin J, Xu X, Li L, Zhang C. cGAS/STING in skin melanoma: from molecular mechanisms to therapeutics. Cell Commun Signal 2024; 22:553. [PMID: 39558334 PMCID: PMC11571982 DOI: 10.1186/s12964-024-01860-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/29/2024] [Indexed: 11/20/2024] Open
Abstract
Melanoma, recognized as the most aggressive type of skin cancer, has experienced a notable increase in cases, especially within populations with fair skin. This highly aggressive cancer is largely driven by UV radiation exposure, resulting in the uncontrolled growth and malignant transformation of melanocytes. The cGAS-STING pathway, an immune signaling mechanism responsible for detecting double-stranded DNA in the cytoplasm, is essential for mediating the immune response against melanoma. This pathway serves a dual purpose: it enhances antitumor immunity by activating immune cells, but it can also promote tumor growth when chronically activated by creating an immunosuppressive environment. This review comprehensively examines the multifaceted implication of the cGAS-STING pathway in melanoma pathogenesis and treatment. We explore its molecular mechanisms, including epigenetic regulation, interaction with signaling pathways such as AR signaling, and modulation by various cellular effectors like TG2 and activin-A. The therapeutic potential of modulating the cGAS-STING pathway is highlighted, with promising results from STING agonists, combination therapies with immune checkpoint inhibitors, and novel drug delivery systems, including nanoparticles and synthetic drugs. Our findings underscore the importance of the cGAS-STING pathway in melanoma, presenting it as a critical target for enhancing anti-tumor immunity. By leveraging this pathway, future therapeutic strategies can potentially convert 'cold' tumors into 'hot' tumors, making them more susceptible to immune responses.
Collapse
Affiliation(s)
- Jafaridarabjerdi Mahin
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xuezhu Xu
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Ling Li
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
3
|
Li W, Guo F, Zeng R, Liang H, Wang Y, Xiong W, Wu H, Yang C, Jin X. CDK4/6 Alters TBK1 Phosphorylation to Inhibit the STING Signaling Pathway in Prostate Cancer. Cancer Res 2024; 84:2588-2606. [PMID: 38861362 DOI: 10.1158/0008-5472.can-23-3704] [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: 11/27/2023] [Revised: 04/02/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
The efficacy of immunotherapy in patients with prostate cancer is limited due to the "cold" tumor microenvironment and the paucity of neoantigens. The STING-TBK1-IRF3 signaling axis is involved in innate immunity and has been increasingly recognized as a candidate target for cancer immunotherapy. Here, we found that treatment with CDK4/6 inhibitors stimulates the STING pathway and enhances the antitumor effect of STING agonists in prostate cancer. Mechanistically, CDK4/6 phosphorylated TBK1 at S527 to inactivate the STING signaling pathway independent of RB1 in prostate cancer cells. CDK4/6-mediated phosphorylation of RB1 at S249/T252 also induced the interaction of RB1 with TBK1 to diminish the phosphorylation of TBK1 at S172, which suppressed STING pathway activation. Overall, this study showed that CDK4/6 suppresses the STING pathway through RB1-dependent and RB1-independent pathways, indicating that CDK4/6 inhibition could be a potential strategy to overcome immunosuppression in prostate cancer. Significance: Inhibiting CDK4/6 activates STING-TBK1-IRF3 signaling in prostate cancer by regulating TBK1 phosphorylation, suggesting that the combination of CDK4/6 inhibitors and STING agonists could be an effective approach to stimulate innate immunity.
Collapse
Affiliation(s)
- Wei Li
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijiang Zeng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Huaiyuan Liang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Wei Xiong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| |
Collapse
|
4
|
Ziegler DV, Parashar K, Fajas L. Beyond cell cycle regulation: The pleiotropic function of CDK4 in cancer. Semin Cancer Biol 2024; 98:51-63. [PMID: 38135020 DOI: 10.1016/j.semcancer.2023.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/02/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023]
Abstract
CDK4, along with its regulatory subunit, cyclin D, drives the transition from G1 to S phase, during which DNA replication and metabolic activation occur. In this canonical pathway, CDK4 is essentially a transcriptional regulator that acts through phosphorylation of retinoblastoma protein (RB) and subsequent activation of the transcription factor E2F, ultimately triggering the expression of genes involved in DNA synthesis and cell cycle progression to S phase. In this review, we focus on the newly reported functions of CDK4, which go beyond direct regulation of the cell cycle. In particular, we describe the extranuclear roles of CDK4, including its roles in the regulation of metabolism, cell fate, cell dynamics and the tumor microenvironment. We describe direct phosphorylation targets of CDK4 and decipher how CDK4 influences these physiological processes in the context of cancer.
Collapse
Affiliation(s)
- Dorian V Ziegler
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Kanishka Parashar
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Lluis Fajas
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; INSERM, Montpellier, France.
| |
Collapse
|
5
|
Singh N, Khan FM, Bala L, Vera J, Wolkenhauer O, Pützer B, Logotheti S, Gupta SK. Logic-based modeling and drug repurposing for the prediction of novel therapeutic targets and combination regimens against E2F1-driven melanoma progression. BMC Chem 2023; 17:161. [PMID: 37993971 PMCID: PMC10666365 DOI: 10.1186/s13065-023-01082-2] [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: 06/12/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Melanoma presents increasing prevalence and poor outcomes. Progression to aggressive stages is characterized by overexpression of the transcription factor E2F1 and activation of downstream prometastatic gene regulatory networks (GRNs). Appropriate therapeutic manipulation of the E2F1-governed GRNs holds the potential to prevent metastasis however, these networks entail complex feedback and feedforward regulatory motifs among various regulatory layers, which make it difficult to identify druggable components. To this end, computational approaches such as mathematical modeling and virtual screening are important tools to unveil the dynamics of these signaling networks and identify critical components that could be further explored as therapeutic targets. Herein, we integrated a well-established E2F1-mediated epithelial-mesenchymal transition (EMT) map with transcriptomics data from E2F1-expressing melanoma cells to reconstruct a core regulatory network underlying aggressive melanoma. Using logic-based in silico perturbation experiments of a core regulatory network, we identified that simultaneous perturbation of Protein kinase B (AKT1) and oncoprotein murine double minute 2 (MDM2) drastically reduces EMT in melanoma. Using the structures of the two protein signatures, virtual screening strategies were performed with the FDA-approved drug library. Furthermore, by combining drug repurposing and computer-aided drug design techniques, followed by molecular dynamics simulation analysis, we identified two potent drugs (Tadalafil and Finasteride) that can efficiently inhibit AKT1 and MDM2 proteins. We propose that these two drugs could be considered for the development of therapeutic strategies for the management of aggressive melanoma.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Lakshmi Bala
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Leibniz Institute for Food Systems Biology, Technical University of Munich, Munich, Germany
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch, South Africa
| | - Brigitte Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, Athens, Greece
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany.
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India.
| |
Collapse
|
6
|
Wang T, Liu W, Shen Q, Tao R, Li C, Shen Q, Lin Y, Huang Y, Yang L, Xie G, Bai J, Li R, Wang L, Tao K, Yin Y. Combination of PARP inhibitor and CDK4/6 inhibitor modulates cGAS/STING-dependent therapy-induced senescence and provides "one-two punch" opportunity with anti-PD-L1 therapy in colorectal cancer. Cancer Sci 2023; 114:4184-4201. [PMID: 37702298 PMCID: PMC10637067 DOI: 10.1111/cas.15961] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Although PARP inhibitor (PARPi) has been proven to be a promising anticancer drug in cancer patients harboring BRCA1/2 mutation, it provides limited clinical benefit in colorectal cancer patients with a low prevalence of BRCA1/2 mutations. In our study, we found PARPi talazoparib significantly induced cellular senescence via inhibiting p53 ubiquitination and activating p21. Furthermore, CDK4/6i palbociclib amplified this therapy-induced senescence (TIS) in vitro and in vivo. Mechanistically, talazoparib and palbociclib combination induced senescence-associated secretory phenotype (SASP), and characterization of SASP components revealed type I interferon (IFN)-related mediators, which were amplified by cGAS/STING signaling. More importantly, RNA sequencing data indicated that combination therapy activated T cell signatures and combination treatment transformed the tumor microenvironment (TME) into a more antitumor state with increased CD8 T cells and natural killer (NK) cells and decreased macrophages and granulocytic myeloid-derived suppressor cells (G-MDSCs). Moreover, clearance of the TIS cells by αPD-L1 promoted survival in immunocompetent mouse colorectal cancer models. Collectively, we elucidated the synergistic antitumor and immunomodulatory mechanisms of the talazoparib-palbociclib combination. Further combination with PD-L1 antibody might be a promising "one-two punch" therapeutic strategy for colorectal cancer patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Gastrointestinal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qian Shen
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ruikang Tao
- Center for Biomolecular Science and EngineeringUniversity of California Santa CruzSanta CruzCaliforniaUSA
| | - Chengguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qian Shen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yao Lin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yongzhou Huang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lei Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gengchen Xie
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jie Bai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lulu Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of PharmacyTianjin Medical UniversityTianjinChina
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
7
|
McRee SK, Bayer AL, Pietruska J, Tsichlis PN, Hinds PW. AKT2 Loss Impairs BRAF-Mutant Melanoma Metastasis. Cancers (Basel) 2023; 15:4958. [PMID: 37894325 PMCID: PMC10605002 DOI: 10.3390/cancers15204958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Despite recent advances in treatment, melanoma remains the deadliest form of skin cancer due to its highly metastatic nature. Melanomas harboring oncogenic BRAFV600E mutations combined with PTEN loss exhibit unrestrained PI3K/AKT signaling and increased invasiveness. However, the contribution of different AKT isoforms to melanoma initiation, progression, and metastasis has not been comprehensively explored, and questions remain about whether individual isoforms play distinct or redundant roles in each step. We investigate the contribution of individual AKT isoforms to melanoma initiation using a novel mouse model of AKT isoform-specific loss in a murine melanoma model, and we investigate tumor progression, maintenance, and metastasis among a panel of human metastatic melanoma cell lines using AKT isoform-specific knockdown studies. We elucidate that AKT2 is dispensable for primary tumor formation but promotes migration and invasion in vitro and metastatic seeding in vivo, whereas AKT1 is uniquely important for melanoma initiation and cell proliferation. We propose a mechanism whereby the inhibition of AKT2 impairs glycolysis and reduces an EMT-related gene expression signature in PTEN-null BRAF-mutant human melanoma cells to limit metastatic spread. Our data suggest that the elucidation of AKT2-specific functions in metastasis might inform therapeutic strategies to improve treatment options for melanoma patients.
Collapse
Affiliation(s)
- Siobhan K. McRee
- Program in Genetics, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA;
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Abraham L. Bayer
- Program in Immunology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA;
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Jodie Pietruska
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Philip N. Tsichlis
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA;
| | - Philip W. Hinds
- Program in Genetics, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA;
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA;
| |
Collapse
|
8
|
McRee SK, Bayer AL, Pietruska J, Tsichlis PN, Hinds PW. AKT2 Loss Impairs BRAF-Mutant Melanoma Metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554685. [PMID: 37662310 PMCID: PMC10473698 DOI: 10.1101/2023.08.24.554685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Despite recent advances in treatment, melanoma remains the deadliest form of skin cancer, due to its highly metastatic nature. Melanomas harboring oncogenic BRAF V600E mutations combined with PTEN loss exhibit unrestrained PI3K/AKT signaling and increased invasiveness. However, the contribution of different AKT isoforms to melanoma initiation, progression, and metastasis has not been comprehensively explored, and questions remain whether individual isoforms play distinct or redundant roles in each step. We investigate the contribution of individual AKT isoforms to melanoma initiation using a novel mouse model of AKT isoform-specific loss in a murine melanoma model, and investigate tumor progression, maintenance, and metastasis among a panel of human metastatic melanoma cell lines using AKT-isoform specific knockdown studies. We elucidate that AKT2 is dispensable for primary tumor formation but promotes migration and invasion in vitro and metastatic seeding in vivo , while AKT1 is uniquely important for melanoma initiation and cell proliferation. We propose a mechanism whereby inhibition of AKT2 impairs glycolysis and reduces an EMT-related gene expression signature in PTEN-null BRAF-mutant human melanoma cells to limit metastatic spread. Our data suggest that elucidation of AKT2-specific functions in metastasis could inform therapeutic strategies to improve treatment options for melanoma patients.
Collapse
|
9
|
Liu J, Zheng R, Zhang Y, Jia S, He Y, Liu J. The Cross Talk between Cellular Senescence and Melanoma: From Molecular Pathogenesis to Target Therapies. Cancers (Basel) 2023; 15:cancers15092640. [PMID: 37174106 PMCID: PMC10177054 DOI: 10.3390/cancers15092640] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Melanoma is a malignant skin tumor that originates from melanocytes. The pathogenesis of melanoma involves a complex interaction that occurs between environmental factors, ultraviolet (UV)-light damage, and genetic alterations. UV light is the primary driver of the skin aging process and development of melanoma, which can induce reactive oxygen species (ROS) production and the presence of DNA damage in the cells, and results in cell senescence. As cellular senescence plays an important role in the relationship that exists between the skin aging process and the development of melanoma, the present study provides insight into the literature concerning the topic at present and discusses the relationship between skin aging and melanoma, including the mechanisms of cellular senescence that drive melanoma progression, the microenvironment in relation to skin aging and melanoma factors, and the therapeutics concerning melanoma. This review focuses on defining the role of cellular senescence in the process of melanoma carcinogenesis and discusses the targeting of senescent cells through therapeutic approaches, highlighting the areas that require more extensive research in the field.
Collapse
Affiliation(s)
- Jiahua Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Runzi Zheng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Yanghuan Zhang
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Shuting Jia
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Yonghan He
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Jing Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|