1
|
Chen Y, Chen H, Han Z, Cui Y, Situ C, Qi Y, Cheng Q, Li Y. PRKCQ Is Dispensable for Spermatogenesis in Mice. Cell Biol Int 2025; 49:522-533. [PMID: 40051302 DOI: 10.1002/cbin.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Protein kinase C (PKC) family is evolutionally conserved and involved in various signaling cascades in all cells. Of the family, PRKCQ is dominatingly expressed in testis, however, its molecular functionality in spermatogenesis and male fertility remains unclear. To evaluate the role of PRKCQ in spermatogenesis, Prkcq knockout mice were generated using CRISPR/Cas9 system. Histological and immunofluorescence assays by different markers were employed to assess the testicular cells variation. Sperm parameters were analyzed by computer-assisted sperm analyzer. qPCR assay was used to examine the expression levels of other PKC family genes. We found that PRKCQ was conserved throughout evolution and highly expressed in testis. Prkcq-/- mice were successfully generated and developed viably. Normal fertility was observed in Prkcq-/- males. Prkcq-/- mice exhibited no defects in spermatogenic cells and mature sperm were full in epididymis. Furthermore, there were no differences in sperm motility and progressive motility between Prkcq-/- males and controls. Our findings report a detailed phenotypic analysis of Prkcq-/- males and indicate that PRKCQ is not required for spermatogenesis in male mice, which can provide basic information for other researchers.
Collapse
Affiliation(s)
- Yu Chen
- Medical Research Center, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Hong Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zhongyan Han
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yiqiang Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Chenghao Situ
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yaling Qi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yan Li
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Qiu M, Zhao L, Li X, Fan Y, Liu M, Hua D, Zhu Y, Liang Y, Zhang Y, Xiao W, Xu X, Li J. Decoding dengue's neurological assault: insights from single-cell CNS analysis in an immunocompromised mouse model. J Neuroinflammation 2025; 22:62. [PMID: 40038739 PMCID: PMC11877810 DOI: 10.1186/s12974-025-03383-w] [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: 12/10/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Dengue encephalitis, a severe neurological complication of dengue virus infection, is increasingly recognized for its rising incidence and significant public health burden. Despite its growing prevalence, the underlying mechanisms and effective therapeutic strategies remain poorly understood. METHODS Cellular atlas of dengue encephalitis was determined by single-nucleus RNA sequencing. Viral load of dengue virus and the level of cytokines expression was detected by RT-qPCR. The target cells of dengue virus were verified by immunofluorescence. The cytotoxic effect of CD8+ T cell was determined by flow cytometry, immunofluorescence, in vivo CD8+ T cell depletion, adoptive transfer and CCK-8-based cell viability assay. Axonal and synaptic reduction induced by dengue virus infection was demonstrated by RT-qPCR, Western blot, transmission electron microscope and immunofluorescence. Finally, motor and sensory functions of mice were detected by open field test and hot plate test, respectively. RESULTS In this study, we utilized single-nucleus RNA sequencing on brain tissues from a dengue-infected murine model to construct a comprehensive cellular atlas of dengue encephalitis. Our findings identify neurons, particularly inhibitory GABAergic subtypes, as the primary targets of dengue virus. Additionally, immune cell infiltration was observed, contributing to significant neurological damage. Comprehensive analyses of cell-cell communication, combined with CD8+ T cell depletion and transfer restoration experiments, have elucidated the critical role of CD8+ T cells in triggering encephalitis through their interaction with neurons. These cells infiltrate the brain from peripheral circulation, interact with neurons, and induce damage of synapse and axon, accompanied by neurological dysfunction. CONCLUSION We defined cellular atlas of dengue encephalitis in mouse model and identified the primary target neuron of dengue virus. In addition, we demonstrated the significant cytotoxic effect of CD8+ T cell, which leads to apoptosis of neuron and neurological dysfunction of mice. Our study provides a molecular and cellular framework for understanding dengue encephalitis through advanced sequencing technologies. The insights gained serve as a foundation for future investigations into its pathogenesis and the development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Minyue Qiu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Lixin Zhao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Xiaojia Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yipei Fan
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Minchi Liu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Dong Hua
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yunkai Zhu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yinyin Liang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Wen Xiao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiaofeng Xu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Jintao Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China.
- Institute of Immunology, Army Medical University, Chongqing, China.
| |
Collapse
|
3
|
Martinez C, Xiong Y, Bartkowski A, Harada I, Ren X, Byerly J, Port E, Jin J, Irie H. A PROTAC degrader suppresses oncogenic functions of PTK6, inducing apoptosis of breast cancer cells. Cell Chem Biol 2025; 32:255-266.e8. [PMID: 39541980 PMCID: PMC11845306 DOI: 10.1016/j.chembiol.2024.10.008] [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: 08/28/2023] [Revised: 06/04/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
Protein tyrosine kinase 6 (PTK6), a non-receptor tyrosine kinase, is an oncogenic driver in many tumor types. However, agents that therapeutically target PTK6 are lacking. Although several PTK6 kinase inhibitors have been developed, none have been clinically translated, which may be due to kinase-independent functions that compromise their efficacy. PTK6 kinase inhibitor treatment phenocopies some, but not all effects of PTK6 downregulation. PTK6 downregulation inhibits growth of breast cancer cells, but treatment with PTK6 kinase inhibitor does not. To chemically downregulate PTK6, we designed a PROTAC, MS105, which potently and specifically degrades PTK6. Treatment with MS105, but not PTK6 kinase inhibitor, inhibits growth and induces apoptosis of breast cancer cells, phenocopying the effects of PTK6 (short hairpin RNA) shRNA/CRISPR. In contrast, both MS105 and PTK6 kinase inhibitor effectively inhibit breast cancer cell migration, supporting the differing kinase dependencies of PTK6's oncogenic functions. Our studies support PTK6 degraders as a preferred approach to targeting PTK6 in cancer.
Collapse
Affiliation(s)
- Criseyda Martinez
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yan Xiong
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alison Bartkowski
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ibuki Harada
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaoxiao Ren
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jessica Byerly
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elisa Port
- Department of Surgery, Mount Sinai Hospital, New York, NY 10029, USA
| | - Jian Jin
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Hanna Irie
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Yue F, Zhao Y, Lv Y, Li S, Wang W, Li Y, Wang S, Wang C. Anti-Tumor Effects of Sheep Umbilical Cord Mesenchymal Stem Cells on Melanoma Cells. Int J Mol Sci 2025; 26:426. [PMID: 39796281 PMCID: PMC11720557 DOI: 10.3390/ijms26010426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
Melanoma is among the most common malignancies and has recently exhibited increased resistance to treatments, resulting in a more aggressive disease course. Mesenchymal stem cells (MSCs) secrete cytokines both in vivo and in vitro, which regulate tumor cell signaling pathways and the tumor microenvironment, thereby influencing tumor progression. This study investigates the anti-melanogenesis effects of sheep umbilical cord mesenchymal stem cells (SUCMSCs) to assess their potential application in melanoma treatment. Our findings indicate that, in vitro, SUCMSCs reduce melanin content and tyrosinase activity, inhibit melanoma cell viability, proliferation, migration, and invasion, and promote melanoma cell apoptosis. Subsequent in vivo experiments confirmed that SUCMSCs effectively suppress tumor growth, and histological analysis via HE staining revealed notable differences. Additionally, transcriptome sequencing analysis indicated that the anti-tumor effects were primarily mediated through autophagy, apoptosis, and the TGF-β and NF-κB signaling pathways. The RT-qPCR validation results aligned with the transcriptome data. In summary, SUCMSCs exert anti-melanogenesis effects through the interaction of multiple signaling pathways and cytokines, demonstrating significant potential for melanoma treatment.
Collapse
Affiliation(s)
- Fengjiao Yue
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yuqing Zhao
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yiting Lv
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Songmei Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Weihai Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Yajun Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Chunsheng Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (F.Y.); (Y.Z.); (Y.L.); (S.L.); (W.W.); (Y.L.)
| |
Collapse
|
5
|
Ye Z, Huang Y, Chen T, Wu Y. Comprehensive analysis of telomere and aging-related signature for predicting prognosis and immunotherapy response in lung adenocarcinoma. J Cardiothorac Surg 2025; 20:31. [PMID: 39757226 DOI: 10.1186/s13019-024-03337-y] [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: 08/28/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a high-risk malignancy. Telomeres- (TRGs) and aging-related genes (ARGs) play an important role in cancer progression and prognosis. This study aimed to develop a novel prognostic model combined TRGs and ARGs signatures to predict the prognosis of patients with LUAD. METHODS LUAD patient's sample data and clinical data were obtained from public databases. The prognostic model was constructed and evaluated using the least absolute shrinkage and selection operator (LASSO), multivariate Cox analysis, time-dependent receiver operating characteristic (ROC), and Kaplan-Meier (K-M) analysis. Immune cell infiltration levels were assessed using single-sample gene set enrichment analysis (ssGSEA). Antitumor drugs with significant correlations between drug sensitivity and the expression of prognostic genes were identified using the CellMiner database. The distribution and expression levels of prognostic genes in immune cells were subsequently analyzed based on the TISCH database. RESULTS This study identified eight characteristic genes that are significantly associated with LUAD prognosis and could serve as independent prognostic factors, with the low-risk group demonstrating a more favorable outcome. Additionally, a comprehensive nomogram was developed, showing a high degree of prognostic predictive value. The results from ssGSEA indicated that the low-risk group had higher immune cell infiltration. Ultimately, our findings revealed that the high-risk group exhibited heightened sensitivity to the Linsitinib, whereas the low-risk group demonstrated enhanced sensitivity to the OSI-027 drug. CONCLUSION The risk score exhibited robust prognostic capabilities, offering novel insights for assessing immunotherapy. This will provide a new direction to achieve personalized and precise treatment of LUAD in the future.
Collapse
Affiliation(s)
- Zhe Ye
- Department of Oncology Radiotherapy, Ruian People's Hospital, Wenzhou Medical University, Affiliated Hospital 3, Wenzhou, Zhejiang, 35200, China
| | - Yiwei Huang
- Department of Oncology Radiotherapy, Ruian People's Hospital, Wenzhou Medical University, Affiliated Hospital 3, Wenzhou, Zhejiang, 35200, China
| | - Tingting Chen
- Department of Oncology Radiotherapy, Ruian People's Hospital, Wenzhou Medical University, Affiliated Hospital 3, Wenzhou, Zhejiang, 35200, China
| | - Youyi Wu
- Department of Oncology Radiotherapy, Ruian People's Hospital, Wenzhou Medical University, Affiliated Hospital 3, Wenzhou, Zhejiang, 35200, China.
- Department of Oncology Radiotherapy, Ruian People's Hospital, Wenzhou Medical University, Affiliated Hospital 3, 108 Ruifeng Avenue, Wenzhou, Zhejiang, 35200, China.
| |
Collapse
|
6
|
Wang Y, Cheng S, Fleishman JS, Chen J, Tang H, Chen ZS, Chen W, Ding M. Targeting anoikis resistance as a strategy for cancer therapy. Drug Resist Updat 2024; 75:101099. [PMID: 38850692 DOI: 10.1016/j.drup.2024.101099] [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: 04/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Anoikis, known as matrix detachment-induced apoptosis or detachment-induced cell death, is crucial for tissue development and homeostasis. Cancer cells develop means to evade anoikis, e.g. anoikis resistance, thereby allowing for cells to survive under anchorage-independent conditions. Uncovering the mechanisms of anoikis resistance will provide details about cancer metastasis, and potential strategies against cancer cell dissemination and metastasis. Here, we summarize the principal elements and core molecular mechanisms of anoikis and anoikis resistance. We discuss the latest progress of how anoikis and anoikis resistance are regulated in cancers. Furthermore, we summarize emerging data on selective compounds and nanomedicines, explaining how inhibiting anoikis resistance can serve as a meaningful treatment modality against cancers. Finally, we discuss the key limitations of this therapeutic paradigm and possible strategies to overcome them. In this review, we suggest that pharmacological modulation of anoikis and anoikis resistance by bioactive compounds could surmount anoikis resistance, highlighting a promising therapeutic regimen that could be used to overcome anoikis resistance in cancers.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Mingchao Ding
- Department of Peripheral Vascular Intervention, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China.
| |
Collapse
|
7
|
Xie J, Ruan S, Tu M, Yuan Z, Hu J, Li H, Li S. Clustering single-cell RNA sequencing data via iterative smoothing and self-supervised discriminative embedding. Oncogene 2024; 43:2279-2292. [PMID: 38834657 DOI: 10.1038/s41388-024-03074-5] [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: 01/21/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Single-cell transcriptome sequencing (scRNA-seq) is a high-throughput technique used to study gene expression at the single-cell level. Clustering analysis is a commonly used method in scRNA-seq data analysis, helping researchers identify cell types and uncover interactions between cells. However, the choice of a robust similarity metric in the clustering procedure is still an open challenge due to the complex underlying structures of the data and the inherent noise in data acquisition. Here, we propose a deep clustering method for scRNA-seq data called scRISE (scRNA-seq Iterative Smoothing and self-supervised discriminative Embedding model) to resolve this challenge. The model consists of two main modules: an iterative smoothing module based on graph autoencoders designed to denoise the data and refine the pairwise similarity in turn to gradually incorporate cell structural features and enrich the data information; and a self-supervised discriminative embedding module with adaptive similarity threshold for partitioning samples into correct clusters. Our approach has shown improved quality of data representation and clustering on seventeen scRNA-seq datasets against a number of state-of-the-art deep learning clustering methods. Furthermore, utilizing the scRISE method in biological analysis against the HNSCC dataset has unveiled 62 informative genes, highlighting their potential roles as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Jinxin Xie
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Shanshan Ruan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Mingyan Tu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhen Yuan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Jianguo Hu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, Shanghai, 200062, China.
- Lingang Laboratory, Shanghai, 200031, China.
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
8
|
Bittman-Soto XS, Thomas ES, Ganshert ME, Mendez-Santacruz LL, Harrell JC. The Transformative Role of 3D Culture Models in Triple-Negative Breast Cancer Research. Cancers (Basel) 2024; 16:1859. [PMID: 38791938 PMCID: PMC11119918 DOI: 10.3390/cancers16101859] [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: 04/19/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Advancements in cell culturing techniques have allowed the development of three-dimensional (3D) cell culture models sourced directly from patients' tissues and tumors, faithfully replicating the native tissue environment. These models provide a more clinically relevant platform for studying disease progression and treatment responses compared to traditional two-dimensional (2D) models. Patient-derived organoids (PDOs) and patient-derived xenograft organoids (PDXOs) emerge as innovative 3D cancer models capable of accurately mimicking the tumor's unique features, enhancing our understanding of tumor complexities, and predicting clinical outcomes. Triple-negative breast cancer (TNBC) poses significant clinical challenges due to its aggressive nature, propensity for early metastasis, and limited treatment options. TNBC PDOs and PDXOs have significantly contributed to the comprehension of TNBC, providing novel insights into its underlying mechanism and identifying potential therapeutic targets. This review explores the transformative role of various 3D cancer models in elucidating TNBC pathogenesis and guiding novel therapeutic strategies. It also provides an overview of diverse 3D cell culture models, derived from cell lines and tumors, highlighting their advantages and culturing challenges. Finally, it delves into live-cell imaging techniques, endpoint assays, and alternative cell culture media and methodologies, such as scaffold-free and scaffold-based systems, essential for advancing 3D cancer model research and development.
Collapse
Affiliation(s)
- Xavier S. Bittman-Soto
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23284, USA; (E.S.T.)
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR 00921, USA
| | - Evelyn S. Thomas
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23284, USA; (E.S.T.)
| | | | | | - J. Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23284, USA; (E.S.T.)
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
9
|
Dai Y, Zhang X, Ou Y, Zou L, Zhang D, Yang Q, Qin Y, Du X, Li W, Yuan Z, Xiao Z, Wen Q. Anoikis resistance--protagonists of breast cancer cells survive and metastasize after ECM detachment. Cell Commun Signal 2023; 21:190. [PMID: 37537585 PMCID: PMC10399053 DOI: 10.1186/s12964-023-01183-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/04/2023] [Indexed: 08/05/2023] Open
Abstract
Breast cancer exhibits the highest global incidence among all tumor types. Regardless of the type of breast cancer, metastasis is a crucial cause of poor prognosis. Anoikis, a form of apoptosis initiated by cell detachment from the native environment, is an outside-in process commencing with the disruption of cytosolic connectors such as integrin-ECM and cadherin-cell. This disruption subsequently leads to intracellular cytoskeletal and signaling pathway alterations, ultimately activating caspases and initiating programmed cell death. Development of an anoikis-resistant phenotype is a critical initial step in tumor metastasis. Breast cancer employs a series of stromal alterations to suppress anoikis in cancer cells. Comprehensive investigation of anoikis resistance mechanisms can inform strategies for preventing and regressing metastatic breast cancer. The present review first outlines the physiological mechanisms of anoikis, elucidating the alterations in signaling pathways, cytoskeleton, and protein targets that transpire from the outside in upon adhesion loss in normal breast cells. The specific anoikis resistance mechanisms induced by pathological changes in various spatial structures during breast cancer development are also discussed. Additionally, the genetic loci of targets altered in the development of anoikis resistance in breast cancer, are summarized. Finally, the micro-RNAs and targeted drugs reported in the literature concerning anoikis are compiled, with keratocin being the most functionally comprehensive. Video Abstract.
Collapse
Affiliation(s)
- Yalan Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Oncology, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Xinyi Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Yingjun Ou
- Clinical Medicine School, Southwest Medicial Univercity, Luzhou, China
- Orthopaedics, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Linglin Zou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Duoli Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingfan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiuju Du
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Li
- Southwest Medical University, Luzhou, China
| | | | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
10
|
Meng J, Zhang C, Zhu N, Zhang C, Liu M, Han Z, Li Y. EPN3 plays oncogenic role in non-small cell lung cancer by activating the JAK1/2-STAT3 pathway. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37186036 DOI: 10.1002/tox.23822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/10/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023]
Abstract
The effect of Epsin 3 (EPN3) on non-small cell lung cancer (NSCLC) has not yet been clearly elucidated. This study identified the exact function of EPN3 on NSCLC progression. EPN3 expression in NSCLC patients were analyzed based on the Cancer Genome Atlas database. Kaplan-Meier analysis was implemented to research the effect of EPN3 on patients' survival. EPN3 expression in clinical tissues of 62 NSCLC cases was monitored by real-time quantitative reverse transcription polymerase chain reaction, immunohistochemistry and Western blot. A549 and H1299 cells were transfected with EPN3 shRNA and treated by RO8191 (20 μM). Proliferation was researched by cell counting kit-8 and 5-ethnyl-2 deoxyuridine assays. Apoptosis was monitored by flow cytometry. Migration and invasion was assessed by Transwell experiment. EPN3 effect on A549 cell in vivo growth was researched using nude mice. RO8191 (200 μg) was intratumoral injected into mice. Immunohistochemistry and Western blot was implemented to monitor protein expression in cells and xenograft tumor tissues. EPN3 was abnormally up-regulated in NSCLC patients and cells, indicating a lower overall survival. Loss of EPN3 weakened proliferation, migration and invasion, induced apoptosis, and repressed epithelial-mesenchymal transition in NSCLC cells. Loss of EPN3 inactivated the JAK1/2-STAT3 pathway in NSCLC cells. RO8191 treatment reversed the inhibition of EPN3 knockdown on the malignant phenotype of NSCLC cells. RO8191 intratumoral injection reversed the suppression of EPN3 silencing on NSCLC cell in vivo growth. EPN3 acted as an oncogene in NSCLC via activating the JAK1/2-STAT3 pathway. EPN3 may be a promising target for NSCLC treatment.
Collapse
Affiliation(s)
- Jiguang Meng
- Department of Pulmonary and Critical Care Medicine, Fourth Medical Center of PLA General Hospital, Beijing, China
- Naval Clinical College, Anhui Medical University, Hefei, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Nengyang Zhu
- Naval Clinical College, Anhui Medical University, Hefei, China
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Caiyun Zhang
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Meng Liu
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yongqun Li
- Department of Pulmonary and Critical Care Medicine, Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Duan A, Zhang Y, Yuan G. Screening of feature genes related to immune and inflammatory responses in periodontitis. BMC Oral Health 2023; 23:234. [PMID: 37085805 PMCID: PMC10122403 DOI: 10.1186/s12903-023-02925-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/30/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Immune and inflammatory responses are important in the occurrence and development of periodontitis. The aim of this study was to screen for immune-related genes and construct a disease diagnostic model to further investigate the underlying molecular mechanisms of periodontitis. METHODS GSE16134 and GSE10334 datasets were used in this study. Differentially expressed genes (DEGs) between the periodontitis and control groups were selected. Immune-related genes were identified, and functional analysis and construction of an interaction network were conducted. Immune characteristics were evaluated using gene set variation analysis GSVA. Immunity-related modules were analyzed using weighted gene co-expression network analysis (WGCNA). The LASSO algorithm was applied to optimize the module genes. Correlation between optimized immune-related DEGs and immune cells was analyzed. RESULTS A total of 324 immune-related DEGs enriched in immune- and inflammation-related functions and pathways were identified. Of which, 23 immune cells were significantly different between the periodontitis and control groups. Nine optimal immune-related genes were selected using the WGCNA and LASSO algorithms to construct a diagnostic model. Except for CXCL1, the other eight genes were significantly positively correlated with regulatory T cells, immature B cells, activated B cells, and myeloid-derived suppressor cells. CONCLUSION This study identified nine immune-related genes and developed a diagnostic model for periodontitis.
Collapse
Affiliation(s)
- Azhu Duan
- Department of Stomatology, Children’s Hospital of Shanghai, Children’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1400 Beijing West Road, Jing’an District, Shanghai, 200000 China
| | - Yeming Zhang
- Department of Stomatology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000 China
| | - Gongjie Yuan
- Department of Stomatology, Children’s Hospital of Shanghai, Children’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1400 Beijing West Road, Jing’an District, Shanghai, 200000 China
| |
Collapse
|
12
|
Bao HJ, Chen X, Liu X, Wu W, Li QH, Xian JY, Zhao Y, Chen S. Box C/D snoRNA SNORD89 influences the occurrence and development of endometrial cancer through 2'-O-methylation modification of Bim. Cell Death Dis 2022; 8:309. [PMID: 35790714 PMCID: PMC9256700 DOI: 10.1038/s41420-022-01102-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022]
Abstract
The small nucleolar RNA (snoRNA) is a type of small non-coding RNA widely distributed in the nucleoli of eukaryotic cells, promoting cancer development. The aim of this study was to assess box C/D snoRNA 89 (SNORD89) dysregulations in endometrial cancer. According to the TCGA database as well as the International Federation of Gynecology and Obstetrics (FIGO), higher SNORD89 expression is found in endometrial cancer tissues. In addition, the SNORD89 expression level was higher in endometrial carcinoma with lymph node metastasis than in endometrial carcinoma without lymph node metastasis. By interacting with the conservative chaperone protein methylase fibrillarin (Fbl), SNORD89 inhibits the translation process of the Bim gene, leading to a decrease in Bim protein. Cancer-promoting effect of SNORD89 can be reversed by Fbl knockdown or Bim overexpressing. What’s more, ASO-mediated silencing of SNORD89 could inhibit endometrial cancer cell proliferation and migration ability. Taken together, SNORD89 can modify Bim through 2′-O-methylation and affect downstream signaling pathways to promote endometrial cancer occurrence and development. The role of methylation modification in the prevention and treatment of endometrial cancer provides a new understanding and SNORD89 may be a new diagnostic and therapeutic target for endometrial cancer. Mechanism of action of SNORD89 ![]()
Collapse
Affiliation(s)
- Hai-Juan Bao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Xi Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Xin Liu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wu Wu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Qian-Hui Li
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jing-Yuan Xian
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shuo Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
13
|
Long Q, Feng L, Li Y, Zuo T, Chang L, Zhang Z, Xu P. Time-resolved quantitative phosphoproteomics reveals cellular responses induced by caffeine and coumarin. Toxicol Appl Pharmacol 2022; 449:116115. [PMID: 35691368 DOI: 10.1016/j.taap.2022.116115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Protein phosphorylation is a critical way that cells respond to external signals and environmental stresses. However, the patterns of cellular response to chemicals at different times were largely unknown. Here, we used quantitative phosphoproteomics to analyze the cellular response of kinases and signaling pathways, as well as pattern change of phosphorylated substrates in HepG2 cells that were exposed to caffeine and coumarin for 10 min and 24 h. Comparing the 10 min and 24 h groups, 33 kinases were co-responded and 32 signaling pathways were co-enriched in caffeine treated samples, while 48 kinases and 34 signaling pathways were co-identified in coumarin treated samples. Instead, the percentage of co-identified phosphorylated substrates only accounted for 4.31% and 9.57% between 10 min and 24 h in caffeine and coumarin treated samples, respectively. The results showed that specific chemical exposure led to a bunch of the same kinases and signaling pathways changed in HepG2 cells, while the phosphorylated substrates were different. In addition, it was found that insulin signaling pathway was significantly enriched by both the caffeine and coumarin treatment. The pattern changes in phosphorylation of protein substrates, kinases and signaling pathways with varied chemicals and different time course shed light on the potential mechanism of cellular responses to endless chemical stimulation.
Collapse
Affiliation(s)
- Qi Long
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Lijie Feng
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China; School of Medicine, Guizhou University, Guiyang 550025, China
| | - Tao Zuo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Zhenpeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China.
| | - Ping Xu
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China; School of Medicine, Guizhou University, Guiyang 550025, China; School of Public Health, China Medical University, Shenyang 110122, China; Hebei Province Key Lab of Research and Application on Microbial Diversity, College of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
14
|
Xu Q, Yu J, Jia G, Li Z, Xiong H. Crocin attenuates NF-κB-mediated inflammation and proliferation in breast cancer cells by down-regulating PRKCQ. Cytokine 2022; 154:155888. [PMID: 35447530 DOI: 10.1016/j.cyto.2022.155888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/06/2022] [Accepted: 04/06/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer confronting women worldwide. Crocin, a glycosylated carotenoid extracted from Crocus sativus L., possesses anti-cancer and anti-inflammatory activities. This study tried to explore the influences of crocin on proliferation and inflammation of BC cells, and to investigate the possible mechanism. The protein levels of protein kinase C theta (PRKCQ) and nuclear factor kappa B (NF-κB) p-p65 and p65 were examined using western blot analysis. The potential targets of crocin were predicted using the PharmMapper database. Cell viability and proliferation were determined utilizing CCK-8 and EdU incorporation assays, respectively. Inflammation was assessed by detecting the levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) using RT-qPCR and ELISA. Results showed that crocin inhibited NF-κB activation and suppressed cell viability and proliferation in BC cells. Crocin caused a significant reduction of levels of TNF-α and IL-1β, suggesting that crocin suppressed inflammation in BC cells. NF-κB inhibition decreased proliferation and inflammation in BC cells. Additionally, PRKCQ was identified as a potential target of crocin according to PharmMapper database. Crocin treatment inhibited the activation of NF-κB in BC cells by reducing PRKCQ expression. Mechanistically, PRKCQ-dependent activation of NF-κB pathway reversed the effects of crocin on the proliferation and inflammation in BC cells. In conclusion, crocin inhibited NF-κB-mediated inflammation and proliferation in BC cells through reducing PRKCQ expression.
Collapse
Affiliation(s)
- Quanxiao Xu
- Department of Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Jinsong Yu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Guangwei Jia
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China.
| | - Zhong Li
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Hui Xiong
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| |
Collapse
|
15
|
Dunn J, McCuaig RD, Tan AHY, Tu WJ, Wu F, Wagstaff KM, Zafar A, Ali S, Diwakar H, Dahlstrom JE, Bean EG, Forwood JK, Tsimbalyuk S, Cross EM, Hardy K, Bain AL, Ahern E, Dolcetti R, Mazzieri R, Yip D, Eastgate M, Malik L, Milburn P, Jans DA, Rao S. Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8 + T Cells in Immunotherapy-Resistant and Metastatic Cancers. Cancers (Basel) 2022; 14:1596. [PMID: 35326747 PMCID: PMC8946217 DOI: 10.3390/cancers14061596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023] Open
Abstract
Protein kinase C (PKC)-θ is a serine/threonine kinase with both cytoplasmic and nuclear functions. Nuclear chromatin-associated PKC-θ (nPKC-θ) is increasingly recognized to be pathogenic in cancer, whereas its cytoplasmic signaling is restricted to normal T-cell function. Here we show that nPKC-θ is enriched in circulating tumor cells (CTCs) in patients with triple-negative breast cancer (TNBC) brain metastases and immunotherapy-resistant metastatic melanoma and is associated with poor survival in immunotherapy-resistant disease. To target nPKC-θ, we designed a novel PKC-θ peptide inhibitor (nPKC-θi2) that selectively inhibits nPKC-θ nuclear translocation but not PKC-θ signaling in healthy T cells. Targeting nPKC-θ reduced mesenchymal cancer stem cell signatures in immunotherapy-resistant CTCs and TNBC xenografts. PKC-θ was also enriched in the nuclei of CD8+ T cells isolated from stage IV immunotherapy-resistant metastatic cancer patients. We show for the first time that nPKC-θ complexes with ZEB1, a key repressive transcription factor in epithelial-to-mesenchymal transition (EMT), in immunotherapy-resistant dysfunctional PD1+/CD8+ T cells. nPKC-θi2 inhibited the ZEB1/PKC-θ repressive complex to induce cytokine production in CD8+ T cells isolated from patients with immunotherapy-resistant disease. These data establish for the first time that nPKC-θ mediates immunotherapy resistance via its activity in CTCs and dysfunctional CD8+ T cells. Disrupting nPKC-θ but retaining its cytoplasmic function may offer a means to target metastases in combination with chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Jenny Dunn
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (J.D.); (R.D.M.); (W.J.T.); (A.L.B.)
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Robert D. McCuaig
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (J.D.); (R.D.M.); (W.J.T.); (A.L.B.)
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Abel H. Y. Tan
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Wen Juan Tu
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (J.D.); (R.D.M.); (W.J.T.); (A.L.B.)
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Fan Wu
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Kylie M. Wagstaff
- Cancer Targeting and Nuclear Therapeutics Lab, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
| | - Anjum Zafar
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Sayed Ali
- Medical Oncology, St John of God Midland Public and Private Hospitals, Perth, WA 6056, Australia;
| | - Himanshu Diwakar
- Woden Specialist Medical Centre, I-MED Radiology Network, Canberra, ACT 2606, Australia;
| | - Jane E. Dahlstrom
- Anatomical Pathology, ACT Pathology, The Canberra Hospital, Canberra Health Services, Canberra, ACT 2605, Australia; (J.E.D.); (E.G.B.)
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra, ACT 0200, Australia; (D.Y.); (L.M.)
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia;
| | - Elaine G. Bean
- Anatomical Pathology, ACT Pathology, The Canberra Hospital, Canberra Health Services, Canberra, ACT 2605, Australia; (J.E.D.); (E.G.B.)
| | - Jade K. Forwood
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia; (J.K.F.); (S.T.); (E.M.C.)
| | - Sofiya Tsimbalyuk
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia; (J.K.F.); (S.T.); (E.M.C.)
| | - Emily M. Cross
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia; (J.K.F.); (S.T.); (E.M.C.)
| | - Kristine Hardy
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| | - Amanda L. Bain
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (J.D.); (R.D.M.); (W.J.T.); (A.L.B.)
| | - Elizabeth Ahern
- Department of Medical Oncology, Monash Health and Monash University, Melbourne, VIC 3168, Australia;
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Riccardo Dolcetti
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (R.D.); (R.M.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Roberta Mazzieri
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (R.D.); (R.M.)
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Desmond Yip
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra, ACT 0200, Australia; (D.Y.); (L.M.)
- Department of Medical Oncology, Canberra Health Services, The Canberra Hospital, Canberra, ACT 2605, Australia
| | - Melissa Eastgate
- Department of Medical Oncology, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia;
- Faculty of Medicine, University of Queensland, Herston, QLD 4006, Australia
| | - Laeeq Malik
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra, ACT 0200, Australia; (D.Y.); (L.M.)
- Department of Medical Oncology, Canberra Health Services, The Canberra Hospital, Canberra, ACT 2605, Australia
| | - Peter Milburn
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia;
| | - David A. Jans
- Nuclear Signaling Lab, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
| | - Sudha Rao
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (J.D.); (R.D.M.); (W.J.T.); (A.L.B.)
- Melanie Swan Memorial Translational Centre, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia; (A.H.Y.T.); (F.W.); (A.Z.); (K.H.)
| |
Collapse
|
16
|
Targeting Protein Kinase C for Cancer Therapy. Cancers (Basel) 2022; 14:cancers14051104. [PMID: 35267413 PMCID: PMC8909172 DOI: 10.3390/cancers14051104] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The protein kinase C (PKC) family belongs to serine-threonine kinases and consists of several subtypes. Increasing evidence suggests that PKCs are critical players in carcinogenesis. Interestingly, PKCs exert both promotive and suppressive effects on tumor cell growth and metastasis, which have attracted immense attention. Herein, we systematically review the current advances in the structure, regulation and biological functions of PKCs, especially the relationship of PKCs with anti-cancer therapy-induced cell death, including the current knowledge of PKCs function in tumor metabolism and microenvironment. Moreover, we discuss the potential role of PKCs as a target for therapeutic intervention in cancer from basic research and clinical trials. Abstract Protein kinase C (PKC) isoforms, a group of serine-threonine kinases, are important regulators in carcinogenesis. Numerous studies have demonstrated that PKC isoforms exert both positive and negative effects on cancer cell demise. In this review, we systematically summarize the current findings on the architecture, activity regulation and biological functions of PKCs, especially their relationship with anti-cancer therapy-induced cell death. Additionally, we elaborate on current knowledge of the effects of PKCs on tumor metabolism and microenvironment, which have gained increasing attention in oncology-related areas. Furthermore, we underscore the basic experimental and clinical implications of PKCs as a target for cancer therapy to evaluate their therapeutic benefits and potential applications.
Collapse
|
17
|
Zheng J, Li G, Wang J, Wang S, Tang Q, Sheng H, Wu W, Wang S. Compound Kushen Injection Protects Skin From Radiation Injury via Regulating Bim. Front Pharmacol 2021; 12:753068. [PMID: 34955827 PMCID: PMC8696473 DOI: 10.3389/fphar.2021.753068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Radiation-induced skin injury is a major side-effect observed in cancer patients who received radiotherapy. Thus identifying new radioprotective drugs for prevention or treatment of post-irradiation skin injury should be prompted. A large number of clinical studies have confirmed that Compound Kushen injection (CKI) can enhance efficacy and reduce toxicity of radiotherapy. The aim of this study is to confirm the effect of CKI in alleviating radiotherapy injury in the skin and explore the exact mechanism. Methods: 60 patients who met the inclusion/exclusion criteria were allocated to treatment group (CKI before radiotherapy) or control group (normal saline before radiotherapy) randomly. MTT assay, flow cytometry, Western Blot, and transient transfection were performed to detect the cell viability, cell apoptosis and Bim expression after treatment with CKI or/and radiotherapy. Results: CKI had the effect of alleviating skin injury in cancer patients who received radiotherapy in clinic. CKI induced cancer cell apoptosis when combined with irradiation (IR), while it reversed the induction of cell apoptosis by IR in human skin fibroblast (HSF) cells. And Bim, as a tumor suppressor, was induced in cancer cells but had no change in HSF cells when treated with CKI. Moreover, the above effect could be attenuated when Bim was silenced by siRNA. Conclusion: We conclude that CKI represents a promising radio-protective agent with a potential differential beneficial effect on both cancer cells (inducing apoptosis) and HSF cells (providing radio-protection via inhibiting IR-induced apoptosis), via regulating Bim. Our study uncovers a novel mechanism by which CKI inhibits human cancer cell while protects skin from radiotherapy, indicating CKI might be a promising radio-protective drug. Clinical Trial Registration: Chinese Clinical Trial Registry (www.chictr.org.cn), identifier ChiCTR2100049164.
Collapse
Affiliation(s)
- Jianxiao Zheng
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Radiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gong Li
- Department of Radiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juanjuan Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shujing Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Tang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honghao Sheng
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanyin Wu
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sumei Wang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
The Anticancer Effects of Flavonoids through miRNAs Modulations in Triple-Negative Breast Cancer. Nutrients 2021; 13:nu13041212. [PMID: 33916931 PMCID: PMC8067583 DOI: 10.3390/nu13041212] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/31/2022] Open
Abstract
Triple- negative breast cancer (TNBC) incidence rate has regularly risen over the last decades and is expected to increase in the future. Finding novel treatment options with minimum or no toxicity is of great importance in treating or preventing TNBC. Flavonoids are new attractive molecules that might fulfill this promising therapeutic option. Flavonoids have shown many biological activities, including antioxidant, anti-inflammatory, and anticancer effects. In addition to their anticancer effects by arresting the cell cycle, inducing apoptosis, and suppressing cancer cell proliferation, flavonoids can modulate non-coding microRNAs (miRNAs) function. Several preclinical and epidemiological studies indicate the possible therapeutic potential of these compounds. Flavonoids display a unique ability to change miRNAs' levels via different mechanisms, either by suppressing oncogenic miRNAs or activating oncosuppressor miRNAs or affecting transcriptional, epigenetic miRNA processing in TNBC. Flavonoids are not only involved in the regulation of miRNA-mediated cancer initiation, growth, proliferation, differentiation, invasion, metastasis, and epithelial-to-mesenchymal transition (EMT), but also control miRNAs-mediated biological processes that significantly impact TNBC, such as cell cycle, immune system, mitochondrial dysregulation, modulating signaling pathways, inflammation, and angiogenesis. In this review, we highlighted the role of miRNAs in TNBC cancer progression and the effect of flavonoids on miRNA regulation, emphasizing their anticipated role in the prevention and treatment of TNBC.
Collapse
|
19
|
Nicolle A, Zhang Y, Belguise K. The Emerging Function of PKCtheta in Cancer. Biomolecules 2021; 11:biom11020221. [PMID: 33562506 PMCID: PMC7915540 DOI: 10.3390/biom11020221] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/30/2022] Open
Abstract
Protein Kinase C theta (PKCθ) is a serine/threonine kinase that belongs to the novel PKC subfamily. In normal tissue, its expression is restricted to skeletal muscle cells, platelets and T lymphocytes in which PKCθ controls several essential cellular processes such as survival, proliferation and differentiation. Particularly, PKCθ has been extensively studied for its role in the immune system where its translocation to the immunological synapse plays a critical role in T cell activation. Beyond its physiological role in immune responses, increasing evidence implicates PKCθ in the pathology of various diseases, especially autoimmune disorders and cancers. In this review, we discuss the implication of PKCθ in various types of cancers and the PKCθ-mediated signaling events controlling cancer initiation and progression. In these types of cancers, the high PKCθ expression leads to aberrant cell proliferation, migration and invasion resulting in malignant phenotype. The recent development and application of PKCθ inhibitors in the context of autoimmune diseases could benefit the emergence of treatment for cancers in which PKCθ has been implicated.
Collapse
|
20
|
Paul A, Singh P, Kuznetsov ML, Karmakar A, Guedes da Silva MFC, Koch B, Pombeiro AJL. Influence of anchoring moieties on new benzimidazole-based Schiff base copper(II) complexes towards estrogen dependent breast cancer cells. Dalton Trans 2021; 50:3701-3716. [PMID: 33634805 DOI: 10.1039/d0dt03873c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two new benzimidazole Schiff base copper(ii) compounds [Cu(5-CH2PPh3-2-salmethylben)(NO3)(H2O)][BF4]·2/3(H2O)·1/3(MeOH) (1) and [Cu(5-CH2NEt3-2-salmethylben)(Cl)][BF4] (2) were synthesised by mixing 2-(1-methyl-1H-benzo[d]imidazol-2-yl)aniline, (3-formyl-4-hydroxybenzyl)triphenylphosphonium chloride or N,N-diethyl-N-(3-formyl-4-hydroxybenzyl)ethanaminium chloride and Cu(NO3)2·3H2O or CuCl2·2H2O in the presence of tetrafluoroborate in a binary mixture of MeOH : H2O under refluxing conditions. The structures of the compounds were established by elemental analysis, FT-IR, ESI-MS analytical techniques and, for 1, by single-crystal X-ray diffraction analysis. Absorption and fluorescence spectroscopic methods were performed to evaluate the calf thymus DNA interactions with the compounds. The calculated binding constants (Kb) of 3.14 × 105 M-1 for 1 and 3.20 × 105 M-1 for 2 were established. The intercalative DNA binding mode was also verified by molecular docking studies. Both compounds demonstrated a notable in vitro cytotoxic effect against human A-549 (lung carcinoma), MCF-7 (breast cancer) and HeLa (cervical cancer) cancer cell lines. A substantial repressive effect on the proliferation of MCF-7 cells (breast cancer cells) was observed for compound 1. The mechanism of action for the effective antiproliferative activity of 1 has additionally been confirmed by means of various biological studies such as morphological assessment through AO/EB, detection of apoptotic induction via Hoechst/PI dual staining, flow cytometry for detection of cell cycle arrest, quantitative analysis of apoptotic cells, DNA degradation, generation of reactive oxygen species (ROS) and by apoptotic induction through mitochondrial staining.
Collapse
Affiliation(s)
- Anup Paul
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Priya Singh
- Departments of Zoology, Faculty of Science, Banaras Hindu University, Varanasi - 221 005, U.P., India.
| | - Maxim L Kuznetsov
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Anirban Karmakar
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - M Fátima C Guedes da Silva
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Biplob Koch
- Departments of Zoology, Faculty of Science, Banaras Hindu University, Varanasi - 221 005, U.P., India.
| | - Armando J L Pombeiro
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal. and Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|