1
|
Shmelev ME, Pilnik AA, Shved NA, Penkova AO, Gulaia VS, Kumeiko VV. IDH1 R132H and TP53 R248Q Mutations Modulate Glioma Cell Migration and Adhesion on Different ECM Components. Int J Mol Sci 2024; 25:12178. [PMID: 39596246 PMCID: PMC11594609 DOI: 10.3390/ijms252212178] [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: 10/22/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Mutations in IDH1 and TP53 have a significant impact on glioma prognosis and progression; however, their roles in tumor cell invasion in terms of interactions with particular components of the extracellular matrix (ECM) are still unclear. Using gene editing protocol based on CRISPR-Cas 9 with cytidine deaminase, we introduced point mutations into U87MG glioblastoma cells to establish modified cell lines with heterozygous IDH1 R132H, homozygous TP53 R248Q and heterozygous IDH1 R132H, homozygous TP53 R248Q genotypes. A comparative study of cell migration on major ECM components was carried out by high-content microscopy. IDH1 R132H mutation introduced to U87MG glioblastoma cells was shown to decrease the migration speed on Matrigel and collagen IV substrates compared to the wild-type. This data were supported by cell adhesion quantification via the lateral shift assay performed by atomic force microscopy (AFM). TP53 R248Q mutation increased cell adhesion to various substrates and significantly promoted cell migration on hyaluronic acid and chondroitin sulfate but did not change the migration rates on laminin and collagens IV and I. A double-mutant genotype produced by consequently introducing IDH1 R132H and TP53 R248Q to parental glioblastoma cells was characterized by the highest migration among all the cell lines, with particularly faster motility on chondroitin sulfate. These findings underscore the complex interactions between glioma cells, with the most important driver mutations and specific ECM components regulating cancer cell migration, offering valuable insights for potential therapeutic targets in glioma treatment.
Collapse
Affiliation(s)
- Mikhail E. Shmelev
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Andrei A. Pilnik
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Nikita A. Shved
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Alina O. Penkova
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Valeriia S. Gulaia
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Vadim V. Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
2
|
Wang C, He Y, Tang J, Mao J, Liang X, Xu M, Zhang Z, Tian J, Jiang J, Li C, Zhou X. Chondroitin sulfate functionalized nanozymes inhibit the inflammation feedback loop for enhanced atherosclerosis therapy by regulating intercellular crosstalk. Int J Biol Macromol 2024; 282:136918. [PMID: 39471920 DOI: 10.1016/j.ijbiomac.2024.136918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
In the inflammatory microenvironment of atherosclerotic plaques, metabolic dysregulation of superoxide anion (O2-) and hydrogen peroxide (H2O2) leads to the activation of feedback mechanisms involving IL-1β, TNF-α, and MCP-1, which triggers inflammatory cascades between macrophages and vascular smooth muscle cells (VSMCs) in atherosclerosis (AS). To address this, a chondroitin sulfate (CS)-functionalized dual-targeted engineered nanozyme, CS-Lip/PB@Rap, was developed by encapsulating mesoporous Prussian blue nanoparticles (PBs) loaded with rapamycin (Rap) within CS-modified liposomes. CS functionalization endowed CS-Lip/PB@Rap with a specific targeting ability for CD44 receptors, thus enabling targeted delivery to inflammatory macrophages and VSMCs. Moreover, its enhanced multiple enzyme-like activities effectively modulated the imbalance of oxidative stress. The underlying mechanism of crosstalk regulation by these engineered nanozymes may inhibit the NF-κB pathway by restoring normal metabolism of O2- and H2O2, thereby blocking the TNF-α, IL-1β, and MCP-1 feedback loops between macrophages and VSMCs. This process reduced the production of inflammatory macrophages and inhibited the VSMC transformation from a contractile phenotype to a synthetic phenotype, preventing the formation of fibrous caps. Furthermore, the elimination of oxidative stress could decrease the production of oxygenized low-density lipoprotein (ox-LDL), which inhibited the formation of foam cells and alleviated the atherogenic progression.
Collapse
Affiliation(s)
- Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingying Mao
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
3
|
Yen H, Liao W, Chen C, Su Y, Huang Y, Hsiao C, Chou Y, Chu Y, Shih P, Liu C. Targeting chondroitin sulfate suppresses macropinocytosis of breast cancer cells by modulating syndecan-1 expression. Mol Oncol 2024; 18:2569-2585. [PMID: 38770553 PMCID: PMC11459036 DOI: 10.1002/1878-0261.13667] [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: 12/26/2023] [Revised: 04/02/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Accumulation of abnormal chondroitin sulfate (CS) chains in breast cancer tissue is correlated with poor prognosis. However, the biological functions of these CS chains in cancer progression remain largely unknown, impeding the development of targeted treatment focused on CS. Previous studies identified chondroitin polymerizing factor (CHPF; also known as chondroitin sulfate synthase 2) is the critical enzyme regulating CS accumulation in breast cancer tissue. We then assessed the association between CHPF-associated proteoglycans (PGs) and signaling pathways in breast cancer datasets. The regulation between CHPF and syndecan 1 (SDC1) was examined at both the protein and RNA levels. Confocal microscopy and image flow cytometry were employed to quantify macropinocytosis. The effects of the 6-O-sulfated CS-binding peptide (C6S-p) on blocking CS functions were tested in vitro and in vivo. Results indicated that the expression of CHPF and SDC1 was tightly associated within primary breast cancer tissue, and high expression of both genes exacerbated patient prognosis. Transforming growth factor beta (TGF-β) signaling was implicated in the regulation of CHPF and SDC1 in breast cancer cells. CHPF supported CS-SDC1 stabilization on the cell surface, modulating macropinocytotic activity in breast cancer cells under nutrient-deprived conditions. Furthermore, C6S-p demonstrated the ability to bind CS-SDC1, increase SDC1 degradation, suppress macropinocytosis of breast cancer cells, and inhibit tumor growth in vivo. Although other PGs may also be involved in CHPF-regulated breast cancer malignancy, this study provides the first evidence that a CS synthase participates in the regulation of macropinocytosis in cancer cells by supporting SDC1 expression on cancer cells.
Collapse
Affiliation(s)
- Hung‐Rong Yen
- Department of Chinese MedicineChina Medical University HospitalTaichungTaiwan
- Chinese Medicine Research Center, and School of Chinese Medicine, College of Chinese MedicineChina Medical UniversityTaichungTaiwan
| | - Wen‐Chieh Liao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- Department of Post‐Baccalaureate Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
| | - Chia‐Hua Chen
- Molecular Medicine Research CenterChang Gung UniversityTaoyuanTaiwan
| | - Ying‐Ai Su
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- College of MedicineChung Shan Medical UniversityTaichungTaiwan
| | - Ying‐Wei Huang
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- College of MedicineChung Shan Medical UniversityTaichungTaiwan
| | - Chi Hsiao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- College of MedicineChung Shan Medical UniversityTaichungTaiwan
| | - Yu‐Lun Chou
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
| | - Yin‐Hung Chu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
| | - Pin‐Keng Shih
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- Department of SurgeryChina Medical University HospitalTaichungTaiwan
- School of MedicineChina Medical UniversityTaichungTaiwan
| | - Chiung‐Hui Liu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
- Department of Post‐Baccalaureate Medicine, College of MedicineNational Chung Hsing UniversityTaichungTaiwan
| |
Collapse
|
4
|
Wang C, Peng J, Xiao Y, Zhang Z, Yang X, Liang X, Yang J, Zhou X, Li C. Advances in nanotherapeutics for tumor treatment by targeting calcium overload. Colloids Surf B Biointerfaces 2024; 245:114190. [PMID: 39232477 DOI: 10.1016/j.colsurfb.2024.114190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Traditional antitumor strategies are facing challenges such as low therapeutic efficacy and high side effects, highlighting the significance of developing non-toxic or low-toxic alternative therapies. As a second messenger, calcium ion (Ca2+) plays an important role in cellular metabolism and communication. However, persistent Ca2+ overload leads to mitochondrial structural and functional dysfunction and ultimately induced apoptosis. Therefore, an antitumor strategy based on calcium overload is a promising alternative. Here, we first reviewed the classification of calcium-based nanoparticles (NPs) for exogenous Ca2+ overload, including calcium carbonate (CaCO3), calcium phosphate (CaP), calcium peroxide (CaO2), and hydroxyapatite (HA), calcium hydroxide, etc. Next, the current endogenous Ca2+ overload strategies were summarized, including regulation of Ca2+ channels, destruction of membrane integrity, induction of abnormal intracellular acidity and oxidative stress. Due to the specificity of the tumor microenvironment, it is difficult to completely suppress tumor development with monotherapy. Therefore, we reviewed the progress based on mitochondrial Ca2+ overload, which improved the treatment efficiency by combining photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), immunogenic cell death (ICD) and gas therapy. We further explored in detail the advantages and promising new targets of this combination antitumor strategies to better address future opportunities and challenges.
Collapse
Affiliation(s)
- Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pharmacy, Yibin Hospital Affiliated to Children's Hospital of Chongqing Medical University, Yibin, Sichuan 644000, China
| | - Junrong Peng
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yiwei Xiao
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Yang
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, China
| | - Xiangyu Zhou
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education, Luzhou, Sichuan 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education, Luzhou, Sichuan 646000, China.
| |
Collapse
|
5
|
Qian W, Zhang L, Zhang F, Ye J, Wan Z, Chen H, Luo C. Integrative analysis reveals the potential prognostic roles and immunological values of unc-5 netrin receptor A (UNC5A) in glioma. Discov Oncol 2024; 15:297. [PMID: 39039366 PMCID: PMC11263319 DOI: 10.1007/s12672-024-01174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 07/17/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND UNC5A had been reported to play crucial roles in multiple cancers. However, little was known about the associations among UNC5A and glioma. Therefore, we first combined scRNA-seq, proteomics, as well as bulk RNA-seq in order to investigate UNC5A's functions in gliomas. METHODS Online databases provided scRNA-seq, proteomics, as well as bulk RNA-seq data on UNC5A in gliomas. The following procedures were conducted in order: QRT-PCR, Norman chart, gene set enrichment analysis (GSEA), and univariate/multifactor Cox regression analyses. We further explored the associations among UNC5A and tumor immunity. RESULTS By comparing gliomas with normal tissues, the TCGA dataset showed a significantly reduced expression of UNC5A, which was also confirmed by GSE50161, GSE4290, and QRT-PCR findings (p < 0.05). In both the TCGA and CGGA datasets, gliomas patients with low-UNC5A expression would have poorer overall survival (OS) prognoses (p < 0.05). ScRNA-seq analysis by the CancerSEA online website presented that UNC5A had a low expression in various glioma clusters and significantly associated with six functional states. Moreover, UNC5A might be a reliable independent biomarker of OS in gliomas patients (p < 0.05). Based on the results of GSEA, UNC5A might be connected to three significant pathways in gliomas. We also successfully created a Norman chart to assess the OS prognoses of these patients. Additionally, in aspects of tumor immunity, the infiltration levels of immune cells in LGG, the immune cell pathways, tumor immune microenvironment, as well as immune checkpoints in both LGG and GBM were revealed to be significantly influenced by UNC5A (p < 0.05). CONCLUSIONS UNC5A was found to have prognostic and immunological significance in gliomas, offering patients with gliomas new treatment options.
Collapse
Affiliation(s)
- Wenbo Qian
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China
| | - Lei Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China
| | - Fenglin Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China
| | - Jingliang Ye
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China
| | - Zhiping Wan
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China
| | - Huairui Chen
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China.
| | - Chun Luo
- Department of Neurosurgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Putuo District, No. 389 Xincun Road, Shanghai, 200092, China.
| |
Collapse
|
6
|
Xu Y, Bai Z, Lan T, Fu C, Cheng P. CD44 and its implication in neoplastic diseases. MedComm (Beijing) 2024; 5:e554. [PMID: 38783892 PMCID: PMC11112461 DOI: 10.1002/mco2.554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/25/2024] Open
Abstract
CD44, a nonkinase single span transmembrane glycoprotein, is a major cell surface receptor for many other extracellular matrix components as well as classic markers of cancer stem cells and immune cells. Through alternative splicing of CD44 gene, CD44 is divided into two isoforms, the standard isoform of CD44 (CD44s) and the variant isoform of CD44 (CD44v). Different isoforms of CD44 participate in regulating various signaling pathways, modulating cancer proliferation, invasion, metastasis, and drug resistance, with its aberrant expression and dysregulation contributing to tumor initiation and progression. However, CD44s and CD44v play overlapping or contradictory roles in tumor initiation and progression, which is not fully understood. Herein, we discuss the present understanding of the functional and structural roles of CD44 in the pathogenic mechanism of multiple cancers. The regulation functions of CD44 in cancers-associated signaling pathways is summarized. Moreover, we provide an overview of the anticancer therapeutic strategies that targeting CD44 and preclinical and clinical trials evaluating the pharmacokinetics, efficacy, and drug-related toxicity about CD44-targeted therapies. This review provides up-to-date information about the roles of CD44 in neoplastic diseases, which may open new perspectives in the field of cancer treatment through targeting CD44.
Collapse
Affiliation(s)
- Yiming Xu
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Chenying Fu
- Laboratory of Aging and Geriatric Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
7
|
Hu ZC, Wang B, Zhou XG, Liang HF, Liang B, Lu HW, Ge YX, Chen Q, Tian QW, Xue FF, Jiang LB, Dong J. Golgi Apparatus-Targeted Photodynamic Therapy for Enhancing Tumor Immunogenicity by Eliciting NLRP3 Protein-Dependent Pyroptosis. ACS NANO 2023; 17:21153-21169. [PMID: 37921421 DOI: 10.1021/acsnano.3c05005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Innate and adaptive immunity is important for initiating and maintaining immune function. The nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome serves as a checkpoint in innate and adaptive immunity, promoting the secretion of pro-inflammatory cytokines and gasdermin D-mediated pyroptosis. As a highly inflammatory form of cell death distinct from apoptosis, pyroptosis can trigger immunogenic cell death and promote systemic immune responses in solid tumors. Previous studies proposed that NLRP3 was activated by translocation to the mitochondria. However, a recent authoritative study has challenged this model and proved that the Golgi apparatus might be a prerequisite for the activation of NLRP3. In this study, we first developed a Golgi apparatus-targeted photodynamic strategy to induce the activation of NLRP3 by precisely locating organelles. We found that Golgi apparatus-targeted photodynamic therapy could significantly upregulate NLRP3 expression to promote the subsequent release of intracellular proinflammatory contents such as IL-1β or IL-18, creating an inflammatory storm to enhance innate immunity. Moreover, this acute NLRP3 upregulation also activated its downstream classical caspase-1-dependent pyroptosis to enhance tumor immunogenicity, triggering adaptive immunity. Pyroptosis eventually led to immunogenic cell death, promoted the maturation of dendritic cells, and effectively activated antitumor immunity and long-lived immune memory. Overall, this Golgi apparatus-targeted strategy provided molecular insights into the occurrence of immunogenic pyroptosis and offered a platform to remodel the tumor microenvironment.
Collapse
Affiliation(s)
- Zhi-Chao Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ben Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Gang Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hai-Feng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bing Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong-Wei Lu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu-Xiang Ge
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Wei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Feng-Feng Xue
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Li-Bo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
8
|
Sun G, Zhao S, Fan Z, Wang Y, Liu H, Cao H, Sun G, Huang T, Cai H, Pan H, Rong D, Gao Y, Tang W. CHSY1 promotes CD8 + T cell exhaustion through activation of succinate metabolism pathway leading to colorectal cancer liver metastasis based on CRISPR/Cas9 screening. J Exp Clin Cancer Res 2023; 42:248. [PMID: 37749638 PMCID: PMC10519095 DOI: 10.1186/s13046-023-02803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 08/19/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND The most common site of metastasis in colorectal cancer (CRC) is the liver and liver metastases occur in more than 50% of patients during diagnosis or treatment. The occurrence of metastasis depends on a series of events known as the invasive-metastasis cascade. Currently, the underlying genes and pathways regulating metastasis initiation in the liver microenvironment are unknown. METHODS We performed systematic CRISPR/Cas9 screening using an in vivo mouse model of CRC liver metastasis to identify key regulators of CRC metastasis. We present the full results of this screen,which included a list of genes that promote or repress CRC liver colonization. By silencing these genes individually, we found that chondroitin sulfate synthase 1 (CHSY1) may be involved in CRC metastasis. We verified the function of CHSY1 and its involvement in liver metastasis of CRC through in vivo and in vitro experiments. RESULT The results of TCGA and CRISPR/Cas9 showed that CHSY1 was overexpressed in CRC primary and liver metastasis tissues and indicated a worse clinical prognosis. In vitro and in vivo experiments confirmed that CHSY1 facilitated the liver metastasis of CRC and CHSY1 induced CD8+ T cell exhaustion and upregulated PD-L1 expression. The metabolomic analysis indicated that CHSY1 promoted CD8+ T cell exhaustion by activating the succinate metabolism pathway leading to liver metastasis of CRC. Artemisinin as a CHSY1 inhibitor reduced liver metastasis and enhanced the effect of anti-PD1 in CRC. PLGA-loaded Artemisinin and ICG probe reduced liver metastasis and increased the efficiency of anti-PD1 treatment in CRC. CONCLUSION CHSY1 could promote CD8+ T cell exhaustion through activation of the succinate metabolic and PI3K/AKT/HIF1A pathway, leading to CRC liver metastasis. The combination of CHSY1 knockdown and anti-PD1 contributes to synergistic resistance to CRC liver metastasis. Artemisinin significantly inhibits CHSY1 activity and in combination with anti-PD1 could synergistically treat CRC liver metastases. This study provides new targets and specific strategies for the treatment of CRC liver metastases, bringing new hope and benefits to patients.
Collapse
Affiliation(s)
- Guangshun Sun
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
- Department of Breast Surgery, the First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhongguo Fan
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yuliang Wang
- School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Hanyuan Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hengsong Cao
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Guoqiang Sun
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Tian Huang
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital &The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Hong Pan
- Department of Breast Surgery, the First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| | - Dawei Rong
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Yun Gao
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Weiwei Tang
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
9
|
Lin YC, Chu YH, Liao WC, Chen CH, Hsiao WC, Ho YJ, Yang MY, Liu CH. CHST11-modified chondroitin 4-sulfate as a potential therapeutic target for glioblastoma. Am J Cancer Res 2023; 13:2998-3012. [PMID: 37559985 PMCID: PMC10408464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 08/11/2023] Open
Abstract
Aberrant chondroitin sulfate (CS) accumulation in glioblastoma (GBM) tissue has been documented, but the role of excessive CS in GBM progression and whether it can be a druggable target are largely unknown. The aim of this study is to clarify the biological functions of CHST11 in GBM cells, and evaluate therapeutic effects of blocking CHST11-derived chondroitin 4-sulfate (C4S). We investigated the expression of CHST11 in glioma tissue by immunohistochemistry, and analyzed CHST11 associated genes using public RNA sequencing datasets. The effects of CHST11 on aggressive cell behaviors have been studied in vitro and in vivo. We demonstrated that CHST11 is frequently overexpressed in GBM tissue, promoting GBM cell mobility and modulating C4S on GBM cells. We further discovered that CSPG4 is positively correlated with CHST11, and CSPG4 involved in CHST11-mediated cell invasiveness. In addition, GBM patients with high expression of CHST11 and CSPG4 have a significantly shorter survival time. We examined the effects of treating C4S-specific binding peptide (C4Sp) as a therapeutic agent in vitro and in vivo. C4Sp treatment attenuated GBM cell invasiveness and, notably, improved survival rate of orthotopic glioma cell transplant mice. Our results propose a possible mechanism of CHST11 in regulating GBM malignancy and highlight a novel strategy for targeting aberrant chondroitin sulfate in GBM cells.
Collapse
Affiliation(s)
- You-Cheng Lin
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Yin-Hung Chu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Wen-Chieh Liao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung UniversityTaoyuan, Taiwan
| | - Wen-Chuan Hsiao
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical UniversityTaichung, Taiwan
| | - Meng-Yin Yang
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General HospitalTaichung, Taiwan
| | - Chiung-Hui Liu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
10
|
Pawlak MR, Smiley AT, Ramirez MP, Kelly MD, Shamsan GA, Anderson SM, Smeester BA, Largaespada DA, Odde DJ, Gordon WR. RAD-TGTs: high-throughput measurement of cellular mechanotype via rupture and delivery of DNA tension probes. Nat Commun 2023; 14:2468. [PMID: 37117218 PMCID: PMC10147940 DOI: 10.1038/s41467-023-38157-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Mechanical forces drive critical cellular processes that are reflected in mechanical phenotypes, or mechanotypes, of cells and their microenvironment. We present here "Rupture And Deliver" Tension Gauge Tethers (RAD-TGTs) in which flow cytometry is used to record the mechanical history of thousands of cells exerting forces on their surroundings via their propensity to rupture immobilized DNA duplex tension probes. We demonstrate that RAD-TGTs recapitulate prior DNA tension probe studies while also yielding a gain of fluorescence in the force-generating cell that is detectable by flow cytometry. Furthermore, the rupture propensity is altered following disruption of the cytoskeleton using drugs or CRISPR-knockout of mechanosensing proteins. Importantly, RAD-TGTs can differentiate distinct mechanotypes among mixed populations of cells. We also establish oligo rupture and delivery can be measured via DNA sequencing. RAD-TGTs provide a facile and powerful assay to enable high-throughput mechanotype profiling, which could find various applications, for example, in combination with CRISPR screens and -omics analysis.
Collapse
Affiliation(s)
- Matthew R Pawlak
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Adam T Smiley
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Maria Paz Ramirez
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Marcus D Kelly
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ghaidan A Shamsan
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sarah M Anderson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | | | | | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wendy R Gordon
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Skeltved N, Nordmaj MA, Berendtsen NT, Dagil R, Stormer EMR, Al-Nakouzi N, Jiang K, Aicher A, Heeschen C, Gustavsson T, Choudhary S, Gögenur I, Christensen JP, Theander TG, Daugaard M, Salanti A, Nielsen MA. Bispecific T cell-engager targeting oncofetal chondroitin sulfate induces complete tumor regression and protective immune memory in mice. J Exp Clin Cancer Res 2023; 42:106. [PMID: 37118819 PMCID: PMC10142489 DOI: 10.1186/s13046-023-02655-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/28/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND The malaria protein VAR2CSA binds oncofetal chondroitin sulfate (ofCS), a unique chondroitin sulfate, expressed on almost all mammalian cancer cells. Previously, we produced a bispecific construct targeting ofCS and human T cells based on VAR2CSA and anti-CD3 (V-aCD3Hu). V-aCD3Hu showed efficacy against xenografted tumors in immunocompromised mice injected with human immune cells at the tumor site. However, the complex effects potentially exerted by the immune system as a result of the treatment cannot occur in mice without an immune system. Here we investigate the efficacy of V-aCD3Mu as a monotherapy and combined with immune checkpoint inhibitors in mice with a fully functional immune system. METHODS We produced a bispecific construct consisting of a recombinant version of VAR2CSA coupled to an anti-murine CD3 single-chain variable fragment. Flow cytometry and ELISA were used to check cell binding capabilities and the therapeutic effect was evaluated in vitro in a killing assay. The in vivo efficacy of V-aCD3Mu was then investigated in mice with a functional immune system and established or primary syngeneic tumors in the immunologically "cold" 4T1 mammary carcinoma, B16-F10 malignant melanoma, the pancreatic KPC mouse model, and in the immunologically "hot" CT26 colon carcinoma model. RESULTS V-aCD3Mu had efficacy as a monotherapy, and the combined treatment of V-aCD3Mu and an immune checkpoint inhibitor showed enhanced effects resulting in the complete elimination of solid tumors in the 4T1, B16-F10, and CT26 models. This anti-tumor effect was abscopal and accompanied by a systemic increase in memory and activated cytotoxic and helper T cells. The combined treatment also led to a higher percentage of memory T cells in the tumor without an increase in regulatory T cells. In addition, we observed partial protection against re-challenge in a melanoma model and full protection in a breast cancer model. CONCLUSIONS Our findings suggest that V-aCD3Mu combined with an immune checkpoint inhibitor renders immunologically "cold" tumors "hot" and results in tumor elimination. Taken together, these data provide proof of concept for the further clinical development of V-aCD3 as a broad cancer therapy in combination with an immune checkpoint inhibitor.
Collapse
Affiliation(s)
- Nanna Skeltved
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mie A Nordmaj
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nicolai T Berendtsen
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Robert Dagil
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emilie M R Stormer
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nader Al-Nakouzi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Ke Jiang
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Christopher Heeschen
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, Candiolo (Torino), Italy
| | - Tobias Gustavsson
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Ismail Gögenur
- Department of Clinical Medicine, University of Copenhagen and Center for Surgical Science, Zealand University Hospital, Copenhagen, Denmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thor G Theander
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mads Daugaard
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Morten A Nielsen
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
12
|
Liu CH, Ho YJ, Wang CY, Hsu CC, Chu YH, Hsu MY, Chen SJ, Hsiao WC, Liao WC. Targeting Chondroitin Sulphate Synthase 1 (Chsy1) Promotes Axon Growth Following Neurorrhaphy by Suppressing Versican Accumulation. Molecules 2023; 28:molecules28093742. [PMID: 37175152 PMCID: PMC10180239 DOI: 10.3390/molecules28093742] [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: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Versican is a chondroitin sulfate proteoglycan (CSPG), which deposits in perineurium as a physical barrier and prevents the growth of axons out of the fascial boundary. Several studies have indicated that the chondroitin sulfate (CS) chains on versican have several possible functions beyond the physical barrier, including the ability to stabilize versican core protein in the extracellular matrix. As chondroitin sulfate synthase 1 (Chsy1) is a crucial enzyme for CS elongation, we hypothesized that in vivo knockdown of Chsy1 at peripheral nerve lesion site may decrease CS and versican accumulation, and result in accelerating neurite regeneration. In the present study, end-to-side neurorrhaphy (ESN) in Wistar rats was used as an in vivo model of peripheral nerve injury to evaluate nerve regeneration after surgical intervention. The distribution and expression of versican and Chsy1 in regenerating axons after ESN was studied using confocal microscopy and western blotting. Chsy1 was silenced at the nerve lesion (surgical) site using in vivo siRNA transfection. The results indicated that Chsy1 was successfully silenced in nerve tissue, and its downregulation was associated with functional recovery of compound muscle action potential. Silencing of Chsy1 also decreased the accumulation of versican core protein, suggesting that transient treating of Chsy1-siRNA may be an alternative and an effective strategy to promote injured peripheral nerve regeneration.
Collapse
Affiliation(s)
- Chiung-Hui Liu
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung 402306, Taiwan
| | - Che-Yu Wang
- School of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan
| | - Chao-Chun Hsu
- School of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan
| | - Yin-Hung Chu
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Min-Yen Hsu
- School of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| | - Shiu-Jau Chen
- Department of Medicine, MacKay Medical College, New Taipei City 252005, Taiwan
- Department of Neurosurgery, MacKay Memorial Hospital, New Taipei City 251020, Taiwan
| | - Wen-Chuan Hsiao
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Wen-Chieh Liao
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| |
Collapse
|
13
|
Kang NW, Visetvichaporn V, Nguyen DT, Shin EK, Kim D, Kim MJ, Yoo SY, Lee JY, Kim DD. Bone tumor-homing nanotherapeutics for prolonged retention in tumor microenvironment and facilitated apoptotic process via mevalonate pathway inhibition. Mater Today Bio 2023; 19:100591. [PMID: 36873733 PMCID: PMC9978036 DOI: 10.1016/j.mtbio.2023.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/12/2023] [Accepted: 02/22/2023] [Indexed: 02/24/2023] Open
Abstract
Bone malignancy features a mineralized extracellular matrix primarily composed of hydroxyapatite, which interferes with the distribution and activity of antineoplastic agents. Herein, we report bone tumor-homing polymeric nanotherapeutics consisting of alendronate-decorated chondroitin sulfate A-graft-poly(lactide-co-glycolide) and doxorubicin (DOX), named PLCSA-AD, which displayed a prolonged retention profile in the tumor microenvironment and augmented therapeutic efficacy via inhibition of the mevalonate pathway. PLCSA-AD exhibited a 1.72-fold lower IC50 value than free DOX and a higher affinity for hydroxyapatite than PLCSA in HOS/MNNG cell-based 2D bone tumor-mimicking models. The inhibition of the mevalonate pathway by PLCSA-AD in tumor cells was verified by investigating the cytosolic fraction of unprenylated proteins, where blank PLCSA-AD significantly increased the expression of cytosolic Ras and RhoA without changing their total cellular amounts. In a bone tumor-mimicking xenografted mouse model, AD-decorated nanotherapeutics significantly increased tumor accumulation (1.73-fold) compared with PLCSA, and higher adsorption to hydroxyapatites was observed in the histological analysis of the tumor. As a result, inhibition of the mevalonate pathway and improvement in tumor accumulation led to markedly enhanced therapeutic efficacy in vivo, suggesting that PLCSA-AD could be promising nanotherapeutics for bone tumor treatment.
Collapse
Affiliation(s)
- Nae-Won Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Voradanu Visetvichaporn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Duy-Thuc Nguyen
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun Kyung Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dahan Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Jae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - So-Yeol Yoo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jae-Young Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
14
|
Natural and synthetic compounds for glioma treatment based on ROS-mediated strategy. Eur J Pharmacol 2023:175537. [PMID: 36871663 DOI: 10.1016/j.ejphar.2023.175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023]
Abstract
Glioma is the most frequent and most malignant tumor of the central nervous system (CNS),accounting for about 50% of all CNS tumor and approximately 80% of the malignant primary tumors in the CNS. Patients with glioma benefit from surgical resection, chemo- and radio-therapy. However these therapeutical strategies do not significantly improve the prognosis, nor increase survival rates owing to restricted drug contribution in the CNS and to the malignant characteristics of glioma. Reactive oxygen species (ROS) are important oxygen-containing molecules that regulate tumorigenesis and tumor progression. When ROS accumulates to cytotoxic levels, this can lead to anti-tumor effects. Multiple chemicals used as therapeutic strategies are based on this mechanism. They regulate intracellular ROS levels directly or indirectly, resulting in the inability of glioma cells to adapt to the damage induced by these substances. In the current review, we summarize the natural products, synthetic compounds and interdisciplinary techniques used for the treatment of glioma. Their possible molecular mechanisms are also presented. Some of them are also used as sensitizers: they modulate ROS levels to improve the outcomes of chemo- and radio-therapy. In addition, we summarize some new targets upstream or downstream of ROS to provide ideas for developing new anti-glioma therapies.
Collapse
|
15
|
Li H, Wang Y, Zhao P, Guo L, Huang L, Li X, Gao W. Naturally and chemically acetylated polysaccharides: Structural characteristics, synthesis, activities, and applications in the delivery system: A review. Carbohydr Polym 2023; 313:120746. [PMID: 37182931 DOI: 10.1016/j.carbpol.2023.120746] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Acetylated polysaccharides refer to polysaccharides containing acetyl groups on sugar units. In the past, the acetylation modification of wall polysaccharides has been a hot research topic for scientists. However, in recent years, many studies have reported that acetylation-modified plant, animal, and microbial polysaccharide show great potential in delivery systems. From the latest perspective, this review systematically presents the different sources of naturally acetylated polysaccharides, the regularity of their modification, the chemical preparation of acetylation modifications, the biological activities and functions of acetylated polysaccharides, and the application in the delivery system. In nature, acetylated polysaccharides are extensively distributed in plants, microorganism, and animals. The level of acetylation modification, the distribution of chains, and the locations of acetylation modification sites differ between species. An increasing number of acetylated polysaccharides were prepared in the aqueous medium, which is safe, environment friendly, and low-cost. In addition to being necessary for plant growth and development, acetylated polysaccharides have immunomodulatory, antioxidant, and anticancer properties. The above-mentioned multiple sources, multifunctional and multi-active acetylated polysaccharides, make them an increasingly important part of delivery systems. We conclude by discussing the future directions for research and development and the potential uses for acetylated polysaccharides.
Collapse
|
16
|
Yang X, Wang Y, Zhao J, Rong H, Chen Y, Xiong M, Ye X, Yu S, Hu H. Coordinated regulation of BACH1 and mitochondrial metabolism through tumor-targeted self-assembled nanoparticles for effective triple negative breast cancer combination therapy. Acta Pharm Sin B 2022; 12:3934-3951. [PMID: 36213532 PMCID: PMC9532561 DOI: 10.1016/j.apsb.2022.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 11/29/2022] Open
Abstract
The poor prognosis of triple negative breast cancer (TNBC) results from a lack of approved targeted therapies coupled with aggressive proliferation and metastasis, which is associated with high recurrence and short overall survival. Here we developed a strategy by employing tumor-targeted self-assembled nanoparticles to coordinately regulate BACH1 (BTB domain and CNC homology 1) and mitochondrial metabolism. The BACH1 inhibitor hemin and mitochondria function inhibitor berberine derivative (BD) were used to prepare nanoparticles (BH NPs) followed by the modification of chondroitin sulfate (CS) on the surface of BH NPs to achieve tumor targeting (CS/BH NPs). CS/BH NPs were found to be able to inhibit tumor migration and invasion by significantly decreasing the amounts of tumor cell metabolites, glycolysis and metastasis-associated proteins, which were related to the inhibition of BACH1 function. Meanwhile, decreased mitochondrial membrane potential, activated caspase 3/9 and increased ROS production demonstrated coordinated regulation of BACH1 and mitochondrial metabolism. In a xenograft mice model of breast cancer, CS/BH NPs significantly inhibited tumor growth and metastasis due to the synergetic effect of hemin and BD without showing obvious toxicities for major organs. In sum, the results of efficacy and safety experiments suggest potential clinical significance of the prepared self-assembled CS/BH nanoparticles for the treatment of TNBC.
Collapse
Affiliation(s)
- Xuan Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yalong Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junke Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hehui Rong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yujun Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Mengting Xiong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoxing Ye
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecules and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecules and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
17
|
Ahmed T, Flores PC, Pan CC, Ortiz HR, Lee YS, Langlais PR, Mythreye K, Lee NY. EPDR1 is a noncanonical effector of insulin-mediated angiogenesis regulated by an endothelial-specific TGF-β receptor complex. J Biol Chem 2022; 298:102297. [PMID: 35872017 PMCID: PMC9396412 DOI: 10.1016/j.jbc.2022.102297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023] Open
Abstract
Insulin signaling in blood vessels primarily functions to stimulate angiogenesis and maintain vascular homeostasis through the canonical PI3K and MAPK signaling pathways. However, angiogenesis is a complex process coordinated by multiple other signaling events. Here, we report a distinct crosstalk between the insulin receptor and endoglin/activin receptor-like kinase 1 (ALK1), an endothelial cell-specific TGF-β receptor complex essential for angiogenesis. While the endoglin-ALK1 complex normally binds to TGF-β or bone morphogenetic protein 9 (BMP9) to promote gene regulation via transcription factors Smad1/5, we show that insulin drives insulin receptor oligomerization with endoglin-ALK1 at the cell surface to trigger rapid Smad1/5 activation. Through quantitative proteomic analysis, we identify ependymin-related protein 1 (EPDR1) as a major Smad1/5 gene target induced by insulin but not by TGF-β or BMP9. We found endothelial EPDR1 expression is minimal at the basal state but is markedly enhanced upon prolonged insulin treatment to promote cell migration and formation of capillary tubules. Conversely, we demonstrate EPDR1 depletion strongly abrogates these angiogenic effects, indicating that EPDR1 is a crucial mediator of insulin-induced angiogenesis. Taken together, these results suggest important therapeutic implications for EPDR1 and the TGF-β pathways in pathologic angiogenesis during hyperinsulinemia and insulin resistance.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - Paola Cruz Flores
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - Christopher C. Pan
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Hannah R. Ortiz
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Yeon S. Lee
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Paul R. Langlais
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Karthikeyan Mythreye
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA,For correspondence: Nam Y. Lee; Karthikeyan Mythreye
| | - Nam Y. Lee
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA,Department of Pharmacology, University of Arizona, Tucson, Arizona, USA,Comprehensive Cancer Center, University of Arizona, Tucson, Arizona, USA,For correspondence: Nam Y. Lee; Karthikeyan Mythreye
| |
Collapse
|