1
|
Ultimo A, Jain A, Gomez-Gonzalez E, Alex TS, Moreno-Borrallo A, Jana S, Ghosh S, Ruiz-Hernandez E. Nanotherapeutic Formulations for the Delivery of Cancer Antiangiogenics. Mol Pharm 2025; 22:2322-2349. [PMID: 40184281 PMCID: PMC12056699 DOI: 10.1021/acs.molpharmaceut.4c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/06/2025]
Abstract
Antiangiogenic medications for cancer treatment have generally failed in showing substantial benefits in terms of prolonging life on their own; their effects are noticeable only when combined with chemotherapy. Moreover, treatments based on prolonged antiangiogenics administration have demonstrated to be ineffective in stopping tumor progression. In this scenario, nanotherapeutics can address certain issues linked to existing antiangiogenic treatments. More specifically, they can provide the ability to target the tumor's blood vessels to enhance drug accumulation and manage release, ultimately decreasing undesired side effects. Additionally, they enable the administration of multiple angiogenesis inhibitors at the same time as chemotherapy. Key reports in this field include the design of polymeric nanoparticles, inorganic nanoparticles, vesicles, and hydrogels for loading antiangiogenic substances like endostatin and interleukin-12. Furthermore, nanoformulations have been proposed to efficiently control relevant pro-angiogenic pathways such as VEGF, Tie2/Angiopoietin-1, HIF-1α/HIF-2α, and TGF-β, providing powerful approaches to block tumor growth and metastasis. In this article, we outline a selection of nanoformulations for antiangiogenic treatments for cancer that have been developed in the past ten years.
Collapse
Affiliation(s)
- Amelia Ultimo
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Ayushi Jain
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Elisabet Gomez-Gonzalez
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Thomson Santosh Alex
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Almudena Moreno-Borrallo
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Sukanya Jana
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Shubhrima Ghosh
- Trinity
Translational Medicine Institute, Trinity College Dublin, the University
of Dublin, St. James’s
Hospital, Dublin 8 D08 NHY1, Ireland
- School
of Biological, Health and Sports Sciences, Technological University Dublin, Grangegorman Lower, Dublin 7 D07 ADY7, Ireland
| | - Eduardo Ruiz-Hernandez
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| |
Collapse
|
2
|
Gao B, Lu Y, Lai X, Xu X, Gou S, Yang Z, Gong Y, Yang H. Metabolic reprogramming in hepatocellular carcinoma: mechanisms of immune evasion and therapeutic implications. Front Immunol 2025; 16:1592837. [PMID: 40370433 PMCID: PMC12075234 DOI: 10.3389/fimmu.2025.1592837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with limited treatment options for advanced stages. Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to the harsh tumor microenvironment (TME) and evade immune surveillance. This review involves the role of metabolic reprogramming in HCC, focusing on the dysregulation of glucose, lipid, and amino acid metabolism, and its impact on immune evasion. Key metabolic pathways, such as the Warburg effect, fatty acid synthesis, and glutaminolysis, are discussed, along with their influence on tumor-associated macrophages (TAMs) and immune cell function. Targeting these metabolic alterations presents a promising therapeutic approach to enhance immunotherapy efficacy and improve HCC patient outcomes.
Collapse
Affiliation(s)
- Bocheng Gao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Lu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingyue Lai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuhua Gou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhida Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Li J, Ma C, Tian S, Liu A, Song Q, Wang N, Song Q, Lin L, Sun P, Wang J. Amide proton transfer-weighted imaging combined with multiple models diffusion-weighted imaging of endometrial cancer: correlations between multi-modal MRI parameters and HIF-1α expression. Front Oncol 2025; 15:1556311. [PMID: 40444079 PMCID: PMC12121162 DOI: 10.3389/fonc.2025.1556311] [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: 01/06/2025] [Accepted: 03/31/2025] [Indexed: 06/02/2025] Open
Abstract
Background Hypoxia inducible factor (HIF-1α) is a major transcriptional factor regulating gene expression under hypoxic conditions. HIF-1α expression was closely correlated with the oxygenation status of tumor and could serve as an important biomarker for tumor hypoxia, aggressiveness, or radiation resistance. High expression of HIF-1α contributes to high aggressiveness or poor prognosis of endometrial cancer. Purpose This study aimed to investigate correlations between multimodal MRI parameters (derived from amide proton transfer weighted imaging [APTw], conventional diffusion weighted imaging [DWI], intravoxel incoherent motion [IVIM] imaging and diffusion kurtosis imaging [DKI]) and HIF-1α expression, and to determine whether multimodal MRI can be used for quantitative evaluation of HIF-1α expression. Study type Retrospective. Population A total of 94 patients with EC were examined with 32 cases finally included in the high HIF-1α expression group and 40 cases included in the low expression group according to the exclusion and inclusion criteria. Field Strength/Sequence 3.0T/APTw, DWI, IVIM, and DKI. Assessment The asymmetry of magnetization transfer rate (MTRasym), apparent diffusion coefficient (ADC), pure diffusion coefficient (D), pseudo diffusion coefficient (D*), perfusion fraction (f), mean kurtosis (MK), and mean diffusivity (MD) were calculated from multimodal MRI and compared between HIF-1α high expression and HIF-1α low expression groups. Statistical Test Mann-Whitney U-test; Chi-square test or Fisher exact test; logistic regression analysis; Area under the receiver operating characteristic (ROC) curve (AUC); The Delong test; Pearson or Spearman correlation coefficients. The significance threshold was set at P < 0.05. Result MTRasym, ADC, D, D*, MK and MD values were significantly higher in high HIF-1α expression than in low HIF-1α expression groups, whereas f value was significantly lower in high HIF-1α expression than in low HIF-1α expression groups. The AUC of HIF-1 α expression evaluated by MTRasym, ADC, D, D*, f, MD, MK and their combination were 0.894 (0.740, 0.973), 0.746 (0.568, 0.879), 0.716 (0.528, 0.904), 0.920 (0.772, 0.984), 0.756 (0.578, 0.886), and 0.973 (0.851-1.000), respectively. Multivariate analysis revealed that only f, MK, and MD values were independent predictors for evaluating HIF-1α expression in EC. Conclusion APTw combined with multi-model diffusion imaging can quantitatively evaluate the expression of HIF-1α in EC, and the combination of multiple quantitative parameters can improve the evaluation efficiency.
Collapse
Affiliation(s)
- Jun Li
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Changjun Ma
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| | - Shifeng Tian
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical Image Artificial Intelligence Engineering Technology Research Center, Dalian, Liaoning, China
| | - Ailian Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical Image Artificial Intelligence Engineering Technology Research Center, Dalian, Liaoning, China
- Technology Innovation Center of Hyperpolarized MRI, Dalian, Liaoning, China
| | - Qingling Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical Image Artificial Intelligence Engineering Technology Research Center, Dalian, Liaoning, China
| | - Nan Wang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical Image Artificial Intelligence Engineering Technology Research Center, Dalian, Liaoning, China
| | - Qingwei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical Image Artificial Intelligence Engineering Technology Research Center, Dalian, Liaoning, China
| | - Liangjie Lin
- Philips Health Technology (China) Co., Ltd., Beijing, China
| | - Peng Sun
- Philips Health Technology (China) Co., Ltd., Beijing, China
| | - Jiazheng Wang
- Philips Health Technology (China) Co., Ltd., Beijing, China
| |
Collapse
|
4
|
Du JY, Zhang CT, Li T, Li YP. Targeting hypoxia and angiogenesis in hepatocellular carcinoma: New insights and therapeutic strategies. World J Hepatol 2024; 16:1371-1376. [PMID: 39744195 PMCID: PMC11686538 DOI: 10.4254/wjh.v16.i12.1371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
In this manuscript we comment on the article by Yang et al published recently, focusing on how hepatic angiopoietin-2 (Ang-2) transcription promote the progression of hepatocellular carcinoma (HCC). HCC is one of the most common and lethal malignancies worldwide, especially in regions with high hepatitis B virus infection rates. Ang-2 is a key mediator of angiogenesis and plays a significant role in the progression of chronic liver diseases towards HCC, particularly in the hypoxic microenvironment. This paper reviews the dynamic expression of Ang-2 in hepatocarcinogenesis and its regulation by hypoxia-inducible factor-1α. Furthermore, we discuss Ang-2's potential as an early monitoring biomarker for metastasis, and the therapeutic prospects of silencing hypoxia-inducible factor-1α to downregulate Ang-2 and suppress epithelial-mesenchymal transition in HCC treatment.
Collapse
Affiliation(s)
- Jia-Yi Du
- Laboratory of Epidemiology and Public Health, Yale University School of Public Health, New Haven, CT 06510, United States
| | - Chu-Ting Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ting Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ya-Ping Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| |
Collapse
|
5
|
Chen A, Huang H, Fang S, Hang Q. ROS: A "booster" for chronic inflammation and tumor metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189175. [PMID: 39218404 DOI: 10.1016/j.bbcan.2024.189175] [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: 05/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Reactive oxygen species (ROS) are a group of highly active molecules produced by normal cellular metabolism and play a crucial role in the human body. In recent years, researchers have increasingly discovered that ROS plays a vital role in the progression of chronic inflammation and tumor metastasis. The inflammatory tumor microenvironment established by chronic inflammation can induce ROS production through inflammatory cells. ROS can then directly damage DNA or indirectly activate cellular signaling pathways to promote tumor metastasis and development, including breast cancer, lung cancer, liver cancer, colorectal cancer, and so on. This review aims to elucidate the relationship between ROS, chronic inflammation, and tumor metastasis, explaining how chronic inflammation can induce tumor metastasis and how ROS can contribute to the evolution of chronic inflammation toward tumor metastasis. Interestingly, ROS can have a "double-edged sword" effect, promoting tumor metastasis in some cases and inhibiting it in others. This article also highlights the potential applications of ROS in inhibiting tumor metastasis and enhancing the precision of tumor-targeted therapy. Combining ROS with nanomaterials strategies may be a promising approach to enhance the efficacy of tumor treatment.
Collapse
Affiliation(s)
- Anqi Chen
- Medical College, Yangzhou University, Yangzhou 225009, China
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng 224006, China; Department of Laboratory Medicine, Yancheng Clinical Medical College of Jiangsu University, Yancheng 224006, China
| | - Sumeng Fang
- School of Mathematics, Tianjin University, Tianjin 300350, China
| | - Qinglei Hang
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining 221200, China; Key Laboratory of Jiangsu Province University for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou 225009, China; Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
6
|
Song L, Zhu C, Shi Q, Xia Y, Liang X, Qin W, Ye T, Yang B, Cao X, Xia J, Zhang K. Gelation embolism agents suppress clinical TACE-incited pro-metastatic microenvironment against hepatocellular carcinoma progression. EBioMedicine 2024; 109:105436. [PMID: 39476535 PMCID: PMC11567102 DOI: 10.1016/j.ebiom.2024.105436] [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: 04/09/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Current embolic agents in transcatheter arterial chemoembolization (TACE) of hepatocellular carcinoma (HCC) encounter instability and easy leakage, discounting TACE efficacy with residual HCC. Moreover, clinical TACE aggravates hypoxia and pro-metastatic microenvironments, rendering patients with HCC poor prognosis. METHODS Herein, we developed Zein-based embolic agents that harness water-insoluble but ethanol-soluble Zein to encompass doxorubicin (DOX)-loaded mesoporous hollow MnO2 (HMnO2). The conditions and capacity of HMnO2 to generate reactive oxygen species (ROS) were assayed. Mechanical examinations of Zein-HMnO2@DOX were performed to evaluate its potential as the embolic agent. In vitro experiments were carried out to evaluate the effect of Zein-HMnO2@DOX on HCC. The subcutaneous HCC mouse model and rabbit VX2 HCC model were established to investigate its anti-tumor and anti-metastasis efficacy and explore its potential anti-tumor mechanism. FINDINGS The high adhesion and crosslinking of Zein with HMnO2@DOX impart Zein-HMnO2@DOX with strong mechanical strength to resist deformation and wash-off. Zein gelation and HMnO2 decomposition in response to water and acidic tumor microenvironment, respectively, enable continuous DOX release and Fenton-like reaction for reactive oxygen species (ROS) production and O2 release to execute ROS-enhanced TACE. Consequently, Zein-based embolic agents outperform clinically-used lipiodol to significantly inhibit orthotopic HCC growth. More significantly, O2 release down-regulates hypoxia inducible factor (HIF-1α), vascular endothelial growth factor (VEGF) and glucose transporter protein 1 (GLUT1), which thereby re-programmes TACE-aggravated hypoxic and pro-metastatic microenvironments to repress HCC metastasis towards lung. Mechanistic explorations uncover that such Zein-based TACE agents disrupt oxidative stress, angiogenesis and glycometabolism pathways to inhibit HCC progression. INTERPRETATION This innovative work not only provides a new TACE agent for HCC, but also establishes a new strategy to ameliorate TACE-aggravated hypoxia and metastasis motivation against clinically-common HCC metastasis after TACE operation. FUNDING Excellent Young Science Fund for National Natural Science Foundation of China (82022033); National Natural Science Foundation of China (Grant No. 82373086, 82102761); Major scientific and technological innovation project of Wenzhou Science and Technology Bureau (Grant No. ZY2021009); Shanghai Young Top-Notch Talent.
Collapse
Affiliation(s)
- Li Song
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Chunyan Zhu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China; Department of Stomatology and Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchangzhong Road, Shanghai, 200072, China
| | - Qing Shi
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, No. 2, Xuefu Lane, Wenzhou, 325000, Zhejiang, China
| | - Yuhan Xia
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170, Shensong Road, Shanghai, 200032, China
| | - Xiayi Liang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Wen Qin
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170, Shensong Road, Shanghai, 200032, China
| | - Biwei Yang
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Jinglin Xia
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China; Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, No. 2, Xuefu Lane, Wenzhou, 325000, Zhejiang, China.
| | - Kun Zhang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
7
|
Abumustafa W, Castven D, Sharif-Askari FS, Abi Zamer B, Hamad M, Marquardt JU, Muhammad JS. PRMT5 Mediated HIF1α Signaling and Ras-Related Nuclear Protein as Promising Biomarker in Hepatocellular Carcinoma. BIOLOGY 2024; 13:216. [PMID: 38666828 PMCID: PMC11048327 DOI: 10.3390/biology13040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/24/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024]
Abstract
Protein arginine N-methyltransferase 5 (PRMT5) has been identified as a potential therapeutic target for various cancer types. However, its role in regulating the hepatocellular carcinoma (HCC) transcriptome remains poorly understood. In this study, publicly available databases were employed to investigate PRMT5 expression, its correlation with overall survival, targeted pathways, and genes of interest in HCC. Additionally, we utilized in-house generated NGS data to explore PRMT5 expression in dysplastic nodules compared to hepatocellular carcinoma. Our findings revealed that PRMT5 is significantly overexpressed in HCC compared to normal liver, and elevated expression correlates with poor overall survival. To gain insights into the mechanism driving PRMT5 overexpression in HCC, we analyzed promoter CpG islands and methylation status in HCC compared to normal tissues. Pathway analysis of PRMT5 knockdown in the HCC cells revealed a connection between PRMT5 expression and genes related to the HIF1α pathway. Additionally, by filtering PRMT5-correlated genes within the HIF1α pathway and selecting up/downregulated genes in HCC patients, we identified Ras-related nuclear protein (RAN) as a target associated with overall survival. For the first time, we report that PRMT5 is implicated in the regulation of HIF1A and RAN genes, suggesting the potential prognostic utility of PRMT5 in HCC.
Collapse
Affiliation(s)
- Wafaa Abumustafa
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Darko Castven
- First Medical Department, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Fatemeh Saheb Sharif-Askari
- Research Institute of Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Batoul Abi Zamer
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mawieh Hamad
- Research Institute of Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Jens-Uwe Marquardt
- First Medical Department, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
8
|
Juhász KZ, Hajdú T, Kovács P, Vágó J, Matta C, Takács R. Hypoxic Conditions Modulate Chondrogenesis through the Circadian Clock: The Role of Hypoxia-Inducible Factor-1α. Cells 2024; 13:512. [PMID: 38534356 PMCID: PMC10969332 DOI: 10.3390/cells13060512] [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: 01/15/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimer transcription factor composed of an alpha and a beta subunit. HIF-1α is a master regulator of cellular response to hypoxia by activating the transcription of genes that facilitate metabolic adaptation to hypoxia. Since chondrocytes in mature articular cartilage reside in a hypoxic environment, HIF-1α plays an important role in chondrogenesis and in the physiological lifecycle of articular cartilage. Accumulating evidence suggests interactions between the HIF pathways and the circadian clock. The circadian clock is an emerging regulator in both developing and mature chondrocytes. However, how circadian rhythm is established during the early steps of cartilage formation and through what signaling pathways it promotes the healthy chondrocyte phenotype is still not entirely known. This narrative review aims to deliver a concise analysis of the existing understanding of the dynamic interplay between HIF-1α and the molecular clock in chondrocytes, in states of both health and disease, while also incorporating creative interpretations. We explore diverse hypotheses regarding the intricate interactions among these pathways and propose relevant therapeutic strategies for cartilage disorders such as osteoarthritis.
Collapse
|
9
|
Zhang X, Yu C, Zhao S, Wang M, Shang L, Zhou J, Ma Y. The role of tumor-associated macrophages in hepatocellular carcinoma progression: A narrative review. Cancer Med 2023; 12:22109-22129. [PMID: 38098217 PMCID: PMC10757104 DOI: 10.1002/cam4.6717] [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: 04/21/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 12/31/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, with complex etiology and mechanism, and a high mortality rate. Tumor-associated macrophages (TAMs) are an important part of the HCC tumor microenvironment. Studies in recent years have shown that TAMs are involved in multiple stages of HCC and are related to treatment and prognosis in HCC. The specific mechanisms between TAMs and HCC are gradually being revealed. This paper reviews recent advances in the mechanisms associated with TAMs in HCC, concentrating on an overview of effects of TAMs on drug resistance in HCC and the signaling pathways linked with HCC, providing clues for the treatment and prognosis determination of HCC.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Chao Yu
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Siqi Zhao
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Min Wang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Longcheng Shang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jin Zhou
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Yong Ma
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
10
|
Zeng Y, Song G, Zhang S, Li S, Meng T, Yuan H, Hu F. GSH-Responsive Polymeric Micelles for Remodeling the Tumor Microenvironment to Improve Chemotherapy and Inhibit Metastasis in Breast Cancer. Biomacromolecules 2023; 24:4731-4742. [PMID: 37672635 DOI: 10.1021/acs.biomac.3c00523] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
The tumor microenvironment (TME) of breast cancer is hypoxic, which can promote tumor progression, including invasion and metastasis, and limit the efficacy of anti-tumor treatment. Nitric oxide (NO) can dilate blood vessels, effectively alleviate hypoxia, and regulate the TME, which has the potential to improve the anti-tumor therapeutic efficacy. Here, chitosan (CO) and octadecylamine (ODA) were linked by the disulfide bond, and the LinTT1 peptide was linked onto CO-SS-ODA for targeting tumor cells and endothelial cells in tumors. The NO donor S-nitroso-N-acetylpenicillamine (SNAP) was connected to CO. Doxorubicin (DOX) was encapsulated, and GSH hierarchically responsive polymer micelles (TSCO-SS-ODA/DOX) were constructed for the treatment of breast cancer. The micelles had differently responsive drug release in different GSH concentrations. In endothelial cells, the micelles rapidly responded to release NO. In tumor cells, the disulfide bond rapidly broke and released DOX to effectively kill tumor cells. The disulfide bond was not sensitive to GSH concentration in endothelial cells, which had less release of DOX. The killing effect of the micelles to endothelial cells was much lower than that to tumor cells. The cell selective drug release of the drug delivery systems enabled safe and effective treatment of drugs. TSCO-SS-ODA/DOX, which had the excellent ability to target tumors, can alleviate tumor hypoxia, decrease the infiltration of M2 macrophages in tumors, increase the infiltration of M1 macrophages in tumors, and remodel the TME. Notably, TSCO-SS-ODA/DOX can significantly inhibit the growth of the primary tumor and effectively inhibit tumor metastasis. The drug delivery system provided a potential solution for effectively treating breast cancer.
Collapse
Affiliation(s)
- Yingping Zeng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Sufen Li
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
11
|
Ma W, Zhang X, Lu J, Lv S, Zhang Z, Ma H, Chen Q, Cao W, Zhang X. Transmembrane protein 147, as a potential Sorafenib target, could expedite cell cycle process and confer adverse prognosis in hepatocellular carcinoma. Mol Carcinog 2023; 62:1295-1311. [PMID: 37212496 DOI: 10.1002/mc.23564] [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: 12/27/2022] [Revised: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 05/23/2023]
Abstract
TMEM147 was identified as a core component of ribosome-bound translocon complex at ER/NE. So far, sparse studies reported its expression profiling and oncological implications in hepatocellular carcinoma (HCC) patients. Here we inspected TMEM147 expression levels in HCC cohorts from public databases and tumor tissues. TMEM147 was augmented at transcriptional levels (p < 0.001) and protein levels in HCC patients. A series of bioinformatics tools implemented in R studio were orchestrated in TCGA-LIHC to evaluate the prognostic significance, compile relevant gene clusters, and explore the oncological functions and therapy responses. It is suggested that TMEM147 could predict poor clinical outcomes effectively and independently (p < 0.001, HR = 2.231 for overall survival (OS) vs. p = 0.04, HR = 2.296 for disease-specific survival), and was related to risk factors including advanced histologic tumor grade (p < 0.001), AFP level (p < 0.001) and vascular invasion (p = 0.007). Functional enrichment analyses indicated that TMEM147 was involved in cell cycle, WNT/MAPK signaling pathways and ferroptosis. Expression profiling in HCC cell lines, mouse model, and a clinical trial revealed that TMEM147 was a considerable target and marker for adjuvant therapy in vitro and in vivo. Subsequentially, in vitro wet-lab experimentation authenticated that TMEM147 would be downregulated by Sorafenib administration in hepatoma cells. Lentivirus-mediated overexpression of TMEM147 could promote cell cycle progression from S phase into G2/M phase, and enhance cell proliferation, thus attenuating drug efficacy and sensitivity of Sorafenib. Further explorations into TMEM147 may inspire a fresh perspective to predict clinical outcomes and improve therapeutic efficacy for HCC patients.
Collapse
Affiliation(s)
- Wanshan Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
| | - Xiaoyu Zhang
- Department of Nephrotic, The Fifth People's Hospital of Jinan, Jinan, Shandong, China
| | - Jing Lu
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
| | - Shiyu Lv
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
| | - Zhenyu Zhang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hongxin Ma
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qianqian Chen
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
| | - Wenjing Cao
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
| | - Xiaoning Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong, China
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
12
|
Buczyńska A, Sidorkiewicz I, Kościuszko M, Adamska A, Siewko K, Dzięcioł J, Szumowski P, Myśliwiec J, Szelachowska M, Popławska-Kita A, Krętowski A. Clinical significance of oxidative stress markers as angioinvasion and metastasis indicators in papillary thyroid cancer. Sci Rep 2023; 13:13711. [PMID: 37608150 PMCID: PMC10444813 DOI: 10.1038/s41598-023-40898-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/17/2023] [Indexed: 08/24/2023] Open
Abstract
Angioinvasion remains the important prognostic feature in papillary thyroid cancer (PTC) patients. Literature data indicates several markers that may be associated with oxidative stress and/or angioinvasion. Therefore, we assessed the utility of selected parameters in angioinvasion and metastasis screening in serum of PTC patients. Serum antioxidant capacity (TAC) and sirtuin 3 (SIRT3) levels were decreased (all p < 0.05) and both DNA/RNA oxidative stress damage products (DNA/RNA OSDP) and malondialdehyde (MDA) levels were increased in PTC patients with angioinvasion and metastasis (study group) when compared with PTC patients without these features (all p < 0.01). The highest screening utility in differentiation between angioinvasion and metastasis presence and absence in PTC patients was presented for DNA/RNA OSDP (AUC = 0.71), SIRT3 (AUC = 0.70), and TAC (AUC = 0.67) (all p < 0.05). Our study suggests that peripheral concentration of oxidative stress markers could be useful as angioinvasion and metastasis indicator in PTC patients.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, 15-276, Białystok, Poland.
| | - Iwona Sidorkiewicz
- Clinical Research Support Centre, Medical University of Bialystok, Ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland
| | - Maria Kościuszko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Katarzyna Siewko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Janusz Dzięcioł
- Department of Human Anatomy, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Piotr Szumowski
- Nuclear Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Janusz Myśliwiec
- Nuclear Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Małgorzata Szelachowska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| | - Anna Popławska-Kita
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland.
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Bialystok, 15-276, Białystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276, Białystok, Poland
| |
Collapse
|
13
|
Cheng Z, Qin H, Cao W, He H, Zhang S, Yang Y, Wang Z, Zou X, Wang L, Huang X, Zhou S, Zhang S. Intravoxel incoherent motion imaging used to assess tumor microvascular changes after transarterial chemoembolization in a rabbit VX2 liver tumor model. Front Oncol 2023; 13:1114406. [PMID: 36925931 PMCID: PMC10011620 DOI: 10.3389/fonc.2023.1114406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Purpose To evaluate the correlation between microvascular density (MVD) and intravoxel incoherent motion (IVIM) magnetic resonance imaging (MRI) parameters and the effect of glycolytic flux after transarterial chemoembolization (TACE) in a rabbit VX2 liver tumor. Materials and methods VX2 liver tumor allografts in 15 New Zealand white rabbits were treated with sterile saline (control group, n = 5) or lipiodol-doxorubicin emulsion (experimental group, n = 10). MRI was performed 2 weeks after the procedure to evaluate IVIM parameters, including apparent diffusion coefficient (ADC), pure diffusion coefficient (D), pseudodiffusion coefficient (D*), and perfusion fraction (PF). All animal samples were taken of the tumor and surrounding liver. Immunostaining for CD31, CD34, CD105, and VEGF was used to evaluate MVD. The protein expression of Glut4, HK2, PKM2, LDHA, and MCT1 was determined using western blotting. Pearson correlation tests were used to analyze the relationship between MVD and IVIM parameters. Results D* value in the peritumoral region was negatively correlated with CD34 (r = -0.71, P = 0.01). PF value positively correlated with CD34 (r = 0.68, P = 0.015), CD105 (r = 0.76, P = 0.004) and VEGF (r = 0.72, P = 0.008) in the peritumoral region. Glut4, HK2, PKM2, and MCT1 in the peritumoral regions were higher in the experimental group than in the control group (all P < 0.05). Conclusion IVIM parameters were correlated with MVD in the intratumoral and peritumoral regions after TACE in a rabbit liver tumor model. The angiogenesis reflected by MVD may be related to changes of glycolytic flux.
Collapse
Affiliation(s)
- Zhimei Cheng
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Huanrong Qin
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Wei Cao
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Huizhou He
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Shuling Zhang
- The Affiliated Hospital of Guizhou Medical University & Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yushi Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenmin Wang
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xun Zou
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Lizhou Wang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Xueqing Huang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Shuai Zhang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| |
Collapse
|
14
|
Ouyang X, Shi X, Huang N, Yang Y, Zhao W, Guo W, Huang Y. WDR72 Enhances the Stemness of Lung Cancer Cells by Activating the AKT/HIF-1 α Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:5059588. [PMID: 36385964 PMCID: PMC9663245 DOI: 10.1155/2022/5059588] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Lung cancer is a common malignant tumor with high morbidity and mortality rate. Lung cancer stem cells are crucial in the development of lung cancer. In this study, we investigate WD repeat-containing protein 72 (WDR72) on lung cancer cell stemness and explore its underlying mechanism. METHODS WDR72 expression was investigated in lung cancer tissues and lung cancer stem cells by Western blot and RT-qPCR. The stemness of lung cancer stem cells was verified by the sphere-forming experiment and the abundance of stem cell markers. For the purpose of determining lung cancer stem cell growth, metastasis, and apoptosis, the CCK-8 assay, colony formation, Transwell migration, and flow cytometry were carried out. The ability of tumorigenesis in vivo was explored by xenograft tumor mouse models. RESULTS Up-regulation of WDR72 was found in lung cancer tissues and lung cancer stem cells. WDR72 overexpression significantly activated the AKT/HIF-1α signaling pathway. Application of PI3K/AKT pathway inhibitor LY29004 was able to counteract the impacts of WDR72 upregulation on genes related to stemness, growth, migration, and apoptosis in lung cancer stem cells. The sphere formation of lung cancer stem cells was significantly diminished after inhibiting the AKT/HIF-1α pathway. The promotion of WDR72 overexpression on lung cancer stem cell proliferation and metastasis was also eliminated by LY29004 treatment. CONCLUSION WDR72 activates the AKT/HIF-1α signaling pathway to enhance the stemness of lung cancer stem cells and promote the growth and metastasis of lung cancer.
Collapse
Affiliation(s)
- Xiaoping Ouyang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou 225002, China
| | - Xinlin Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou 225002, China
| | - Na Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Yuping Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Wei Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Wei Guo
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Yumin Huang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou 225002, China
| |
Collapse
|