1
|
Zeng Y, Tao Y, Du G, Huang T, Chen S, Fan L, Zhang N. Advances in the mechanisms of HIF-1α-enhanced tumor glycolysis and its relation to dedifferentiation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 197:1-10. [PMID: 40373959 DOI: 10.1016/j.pbiomolbio.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Metabolic reprogramming, a hallmark of malignancy, enables tumor cells to adapt to the harsh and dynamic tumor microenvironment (TME) by altering metabolic pathways. Hypoxia, prevalent in solid tumors, activates hypoxia inducible factor 1α (HIF-1α). HIF-1α drives metabolic reprogramming, enhancing glycolysis primarily through the Warburg effect to reduce oxygen dependence and facilitate tumor cell growth/proliferation. The above process is associated with accelerated tumor cell dedifferentiation and enhanced stemness, generating cancer stem cells (CSCs) which possesses the potential for self-renewal and differentiation that can differentiate into a wide range of subtypes of tumor cells and fuel tumor heterogeneity, metastasis, and recurrence, complicating therapy. This review examines the HIF-1α-glycolysis-dedifferentiation crosstalk mechanisms, expecting that indirect inhibition of HIF-1α by targeting metabolic enzymes, metabolites, or their signaling pathways will offer an effective therapeutic strategy to improve the cancer treatment outcomes.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yonggang Tao
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guotu Du
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tianyu Huang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shicheng Chen
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Longmei Fan
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Neng Zhang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
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
|
3
|
Monjezi S, Soleimani V, Cheraghzadeh M, Ghanbari S, Adelipour M. CIRBP mRNA level in breast cancer is associated with HIF1α gene expression and microvascular density. BMC Res Notes 2025; 18:202. [PMID: 40320538 PMCID: PMC12049796 DOI: 10.1186/s13104-025-07265-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVE Based on the available evidence, the cold-inducible RNA-binding protein (CIRBP) appears to play a role in increasing the stability of hypoxia-inducible factor 1-alpha (HIF1α) mRNA. This research aimed to examine the levels of CIRBP and HIF1α mRNA within breast tumor tissues and explore the relationship between their gene expression and tumor Microvascular density (MVD). RESULTS The results revealed a significant upregulation of CIRBP (5.07-fold) and HIF1α (4.5-fold) gene expression in BC samples compared to the surrounding normal tissues (p < 0.001). This upregulation was also associated with various clinicopathologic features. Furthermore, there was a correlation between CIRBP mRNA expression, HIF1α mRNA expression, and MVD. Consequently, this study suggests that CIRBP may have a role in promoting angiogenesis in BC.
Collapse
Affiliation(s)
- Sajad Monjezi
- Department of Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and molecular research center, Medical Basic Science Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Soleimani
- Department of Pathology, School of Medicine, Cancer Institute, Tehran University of Medical Science, Tehran, Iran
| | - Maryam Cheraghzadeh
- Department of Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeed Ghanbari
- Department of Biostatistics and Epidemiology, School of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Adelipour
- Department of Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Cellular and molecular research center, Medical Basic Science Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
4
|
Owida HA, Saleh RO, Mohammad SI, Vasudevan A, Roopashree R, Kashyap A, Nanda A, Ray S, Hussein A, Yasin HA. Deciphering the role of circular RNAs in cancer progression under hypoxic conditions. Med Oncol 2025; 42:191. [PMID: 40314834 DOI: 10.1007/s12032-025-02727-z] [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: 02/23/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
Hypoxia, characterized by reduced oxygen levels, plays a pivotal role in cancer progression, profoundly influencing tumor behavior and therapeutic responses. A hallmark of solid tumors, hypoxia drives significant metabolic adaptations in cancer cells, primarily mediated by hypoxia-inducible factor-1α (HIF-1α), a key transcription factor activated in low-oxygen conditions. This hypoxic environment promotes epithelial-mesenchymal transition (EMT), enhancing cancer cell migration, metastasis, and the development of cancer stem cell-like properties, which contribute to therapy resistance. Moreover, hypoxia modulates the expression of circular RNAs (circRNAs), leading to their accumulation in the tumor microenvironment. These hypoxia-responsive circRNAs regulate gene expression and cellular processes critical for cancer progression, making them promising candidates for diagnostic and prognostic biomarkers in various cancers. This review delves into the intricate interplay between hypoxic circRNAs, microRNAs, and RNA-binding proteins, emphasizing their role as molecular sponges that modulate gene expression and signaling pathways involved in cell proliferation, apoptosis, and metastasis. It also explores the relationship between circRNAs and the tumor microenvironment, particularly how hypoxia influences their expression and functional dynamics. Additionally, the review highlights the potential of circRNAs as diagnostic and prognostic tools, as well as their therapeutic applications in innovative cancer treatments. By consolidating current knowledge, this review underscores the critical role of circRNAs in cancer biology and paves the way for future research aimed at harnessing their unique properties for clinical advancements. Specifically, this review examines the biogenesis, expression patterns, and mechanistic actions of hypoxic circRNAs, focusing on their ability to act as molecular sponges for microRNAs and their interactions with RNA-binding proteins. These interactions impact key signaling pathways related to tumor growth, metastasis, and drug resistance, offering new insights into the complex regulatory networks governed by circRNAs under hypoxic stress.
Collapse
Affiliation(s)
- Hamza Abu Owida
- Department of Medical Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman, Jordan
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Suleiman Ibrahim Mohammad
- Research Follower, INTI International University, 71800, Negeri Sembilan, Malaysia.
- Electronic Marketing and Social Media, Economic and Administrative Sciences, Zarqa University, Zarqa, Jordan.
| | - Asokan Vasudevan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ahmed Hussein
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
5
|
Liang Y, Su T, Zhu S, Sun R, Qin J, Yue Z, Wang X, Liang Z, Tan X, Bian Y, Zhao F, Tang D, Yin G. Astragali Radix-Curcumae Rhizoma normalizes tumor blood vessels by HIF-1α to anti-tumor metastasis in colon cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156562. [PMID: 40023968 DOI: 10.1016/j.phymed.2025.156562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Abnormal tumor blood vessels can significantly promote the malignant progression of tumors, prompting researchers to focus on drugs that normalize these vessels for clinical treatment. The combination of the Qi-tonifying drug Astragali Radix and the blood-activating drug Curcumae Rhizoma, referred to as AC, exhibited significant anti-tumor metastasis effects. However, the association between the anti-tumor metastasis effect of AC and its potential role in regulating tumor vascular remodeling warrants further exploration. PURPOSE This study aimed to elucidate the mechanism through which AC induces tumor blood vessel normalization in colon cancer (CC). METHODS The potential active components of AC were identified through UPLC-MS/MS. An orthotopic transplantation model of CC was established in BALB/c mice using the CT26-Lucifer cell line, and the effects of AC were evaluated using IVIS imaging, hematoxylin and eosin (H&E) staining, and immunohistochemistry. Network pharmacology and molecular biology analyses were employed to identify the potential direct targets of AC. Subsequently, RT-PCR and Western blotting techniques were utilized to validate the findings obtained from network pharmacology. Furthermore, ELISA and other methodologies were used to investigate glycolysis-related indicators, along with immunofluorescence technology to demonstrate changes in vascular leakage and perfusion characteristics associated with blood vessel normalization. RESULTS We identified HIF-1α as a potential direct target of AC. This interaction influences the glycolytic processes in both tumor cells and tumor-associated endothelial cells (TECs) by directly binding to HIF-1α and modulating its nuclear translocation, thereby determining the integrity of TEC junctions. Mechanistically, AC directly regulates the key enzyme PFKFB3 in glycolysis by modulating HIF-1α expression and inhibiting its nuclear translocation. This action reduces tumor glycolytic flux, decreases the internalization of VE-cad, and influences the expression of downstream matrix metalloproteinases (MMPs), thereby strengthening the adherens and tight junctions between TECs and restoring vascular integrity. CONCLUSION This study presents novel findings that AC can regulate glycolysis through the inhibition of HIF-1α nuclear translocation, thereby promoting the normalization of tumor blood vessels and effectively inhibiting tumor metastasis. These results suggested that AC may serve as an effective therapeutic agent for normalizing tumor blood vessels.
Collapse
Affiliation(s)
- Yan Liang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingting Su
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijiao Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ruolan Sun
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiahui Qin
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zengyaran Yue
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu Wang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhongqing Liang
- School of Acupuncture-Moxibustion and Tuina · School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiying Tan
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yong Bian
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fan Zhao
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Decai Tang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Gang Yin
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
6
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
7
|
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J, Xiao Y. Targeting ncRNAs to overcome metabolic reprogramming‑mediated drug resistance in cancer (Review). Int J Oncol 2025; 66:35. [PMID: 40116120 PMCID: PMC12002672 DOI: 10.3892/ijo.2025.5741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025] Open
Abstract
The emergence of resistance to antitumor drugs in cancer cells presents a notable obstacle in cancer therapy. Metabolic reprogramming is characterized by enhanced glycolysis, disrupted lipid metabolism, glutamine dependence and mitochondrial dysfunction. In addition to promoting tumor growth and metastasis, metabolic reprogramming mediates drug resistance through diverse molecular mechanisms, offering novel opportunities for therapeutic intervention. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that lack protein‑coding function, represent a notable fraction of the human genome. Due to their distinct expression profiles and multifaceted roles in various cancers, ncRNAs have relevance in cancer pathophysiology. ncRNAs orchestrate metabolic abnormalities associated with drug resistance in cancer cells. The present review provides a comprehensive analysis of the mechanisms by which metabolic reprogramming drives drug resistance, with an emphasis on the regulatory roles of ncRNAs in glycolysis, lipid metabolism, mitochondrial dysfunction and glutamine metabolism. Furthermore, the present review aimed to discuss the potential of ncRNAs as biomarkers for predicting chemotherapy responses, as well as emerging strategies to target ncRNAs that modulate metabolism, particularly in the context of combination therapy with anti‑cancer drugs.
Collapse
Affiliation(s)
- Junxin Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yanyu Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Lin Fu
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Huiling Chen
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital of Southwest Medical University, Meishan, Sichuan 64200, P.R. China
| | - Zhongshu Wang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yu Huang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jidong Miao
- Department of Oncology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yi Xiao
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
8
|
Jin Y, Wang Y, Wang C, Zhang L, Gao D, Liu H, Cao Q, Tian C, Bian Y, Wang Y. Salidroside inhibits osteoclast differentiation based on osteoblast-osteoclast interaction via HIF-1a pathway. Chin J Nat Med 2025; 23:572-584. [PMID: 40383613 DOI: 10.1016/s1875-5364(25)60864-8] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 01/02/2025] [Indexed: 05/20/2025]
Abstract
This study investigated the regulatory potential of salidroside (SAL), a primary active compound in Rhodiola rosea L., on osteoclast differentiation by modulating the hypoxia-inducible factor 1-alpha (HIF-1a) pathway in osteoblasts. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were employed to validate whether the receptor activator of nuclear factor-?B ligand (RANKL) is the downstream target gene of HIF-1a in osteoblasts. The study also utilized lipopolysaccharide (LPS)-induced mouse osteolysis to examine the impact of SAL on osteolysis in vivo. Furthermore, conditioned medium (CM) from SAL-pretreated osteoblasts was used to investigate the paracrine effects on osteoclastogenesis through the HIF-1a pathway. Hypoxic condition-induced overexpression of HIF-1a upregulated RANKL levels by binding to the RANKL promoter and enhancing transcription in osteoblastic cells. In vivo, SAL significantly alleviated bone tissue hypoxia and decreased the expression of HIF-1a by downregulating the expression of RANKL, vascular endothelial growth factor (VEGF), interleukin 6 (IL-6), and angiopoietin-like 4 (ANGPTL4). In the paracrine experiment, conditioned media from SAL-pretreated osteoblasts inhibited differentiation through the HIF-1a/RANKL, VEGF, IL-6, and ANGPTL4 pathways. RANKL emerges as the downstream target gene regulated by HIF-1a in osteoblasts. SAL significantly alleviates bone tissue hypoxia and bone loss in LPS-induced osteolysis through the HIF-1a/RANKL, VEGF, IL-6, and ANGPTL4 pathways. SAL inhibits osteoclast differentiation by regulating osteoblast paracrine secretion.
Collapse
Affiliation(s)
- Yutong Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chuan Wang
- Department of Stomatology, NHC Key Laboratory of Hormones and Development, Chu Hsien- I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Department of Stomatology, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China
| | - Lingling Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Dandan Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haizhao Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qingwen Cao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chenchen Tian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Yue Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
9
|
Jeng LB, Shih FY, Liao YW, Shyu WC, Teng CF. Hypoxic tumor cell line lysate-pulsed dendritic cell vaccine exhibits better therapeutic effects on hepatocellular carcinoma. Br J Cancer 2025; 132:837-848. [PMID: 40050434 PMCID: PMC12041587 DOI: 10.1038/s41416-025-02975-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dendritic cell (DC) vaccine is a promising immunotherapy for hepatocellular carcinoma (HCC) via triggering antigen-specific anti-tumor immunity. Hypoxia contributes to higher level and broader spectrum of antigen expression in tumor cells. METHODS This study aims to compare immunological activity and therapeutic efficacy between hypoxic and normoxic HCC cell line lysate-pulsed DC vaccines. RESULTS The results showed that hypoxic HCC cell line lysate-pulsed DC vaccines exhibited a stronger activity in producing interleukin-12 and promoting T cell proliferation and cytotoxicity in vitro. In HCC mice, hypoxic HCC cell line lysate-pulsed DC vaccines displayed a better efficacy in improving survival time and tumor volume and inducing intratumoral cytotoxic T cell infiltration and activation as well as tumor cell apoptosis. Adenylate kinase 4-derived antigens were important for hypoxic HCC cell line lysate-pulsed DC vaccine-elicited T cell killing. CONCLUSIONS In conclusion, this study demonstrated hypoxic HCC cell line lysate-pulsed DC vaccine as a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan, ROC
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
- School of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, Taiwan, ROC
| | - Yu-Wen Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan, ROC
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Occupational Therapy, Asia University, Taichung, Taiwan, ROC
| | - Chiao-Fang Teng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC.
- Master Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
10
|
Hou YJ, Yang XX, Meng HX. Mitochondrial metabolism in laryngeal cancer: therapeutic mechanisms and prospects. Biochim Biophys Acta Rev Cancer 2025; 1880:189335. [PMID: 40311711 DOI: 10.1016/j.bbcan.2025.189335] [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: 02/02/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
Tumours reprogram pathways that regulate nutrient uptake and metabolism to meet the biosynthetic, bioenergetic, and redox requirements of cancer cells. This phenomenon is known as metabolic reprogramming and is edited by the deletion of oncogenes and the activation of proto-oncogenes. This article highlights the pathological mechanisms associated with metabolic reprogramming in laryngeal cancer (LC), including enhanced glycolysis, tricarboxylic acid cycle, nucleotide synthesis, lipid synthesis and metabolism, and amino acid metabolism, with a special emphasis on glutamine, tryptophan, and arginine metabolism. All these changes are regulated by HPV infection, hypoxia, and metabolic mediators in the tumour microenvironment. We analyzed the function of metabolic reprogramming in the development of drug resistance during standard LC treatment, which is challenging. In addition, we revealed recent advances in targeting metabolic strategies, assessing the strengths and weaknesses of clinical trials and treatment programs to attack resistance. This review summarises some currently important biomarkers and lays the foundation for therapeutic pathways in LC.
Collapse
Affiliation(s)
- Yun-Jing Hou
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin-Xin Yang
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Xue Meng
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
11
|
Cooper DH, Almendros I, Kendzerska T. Sleep, Circadian Rhythms, and Lung Cancer. Semin Respir Crit Care Med 2025. [PMID: 39900110 DOI: 10.1055/a-2531-1059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide, with the prevalence of the disease continually rising. Therefore, identifying disease-modifying risk factors is critical, with increasing recognition of the impact of sleep quality/sleep disorders. This narrative review summarizes the evidence on the role of five domains of sleep on lung cancer incidence and progression: (i) sleep quality/duration, (ii) sleep disordered breathing, (iii) circadian rhythm disturbances, (iv) sleep-related movement disorders, and (v) personal, environmental, and social factors that modulate each of these associations. Epidemiological evidence supports reduced sleep duration, increased sleep duration, poor sleep quality, insomnia, obstructive sleep apnea, evening chronotype, peripheral limb movements in sleep, and less robustly for night shift work and restless leg syndrome to be associated with increased risk of lung cancer development, with potential impacts on cancer survival outcomes. Proposed mechanisms underlying the biological plausibility of these epidemiological associations are also explored, with common theories relating to immune dysregulation, metabolic alterations, reductions in melatonin, sympathetic overactivation, increased reactive oxygen species, production of protumorigenic exosomes, and inflammation. We also summarized potential treatments addressing impaired sleep quality/sleep disorders and their ability to attenuate the risk of lung cancer and improve cancer survival. Although evidence on reversibility is inconsistent, there are trends toward positive outcomes. Future research should focus on clinical trials to confirm cause and effect relationships, large epidemiologic studies for incidence/prognosis, clarification on the relative efficacy of treatment modalities, and more in vivo animal models to establish the molecular mechanisms underlying these relationships.
Collapse
Affiliation(s)
- Daniel H Cooper
- The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Tetyana Kendzerska
- The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
12
|
Zhou RT, Luo XJ, Zhang XXR, Wu JF, Ni YR. The potential of miR-29 in modulating tumor angiogenesis: a comprehensive review. Discov Oncol 2025; 16:474. [PMID: 40189720 PMCID: PMC11973036 DOI: 10.1007/s12672-025-02246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of short non-coding RNAs that play a crucial role in the post-transcriptional regulation of gene expression. They are associated with various biological processes related to tumors. Among the numerous miRNAs, miR-29 has garnered attention for its role in regulating tumor angiogenesis. In numerous human tumors, miR-29 has been demonstrated to negatively correlate with the capacity for angiogenesis and the degree of malignancy, as well as with the expression levels of pro-angiogenic factors such as vascular endothelial growth factor vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and matrix metalloproteinase (MMP)-2. Multiple studies, utilizing techniques like dual-luciferase reporter assays, have confirmed that miR-29 directly targets the 3'-untranslated region (UTR) of mRNAs for VEGF, PDGF, and MMP-2. Extensive investigations involving tumor cell lines and animal models have shown that the overexpression of miR-29, achieved through miRNA transfection or the introduction of miRNA mimics, effectively inhibits angiogenesis by upregulating these pro-angiogenic factors. Conversely, downregulation of miR-29 using specific inhibitors promotes angiogenesis. While small molecule inhibitors and antibodies targeting VEGF constitute a primary strategy in anti-angiogenesis therapies, miR-29's ability to target multiple pro-angiogenic molecules positions it as a promising candidate for future therapeutic interventions, especially with ongoing advancements in nucleic acid drug design and delivery systems.
Collapse
Affiliation(s)
- Rui-Ting Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Department of Gastroenterology, The First College of Clinical Medical Science and Yichang Central People's Hospital, China Three Gorges University, Yichang, 443003, China
- Division of Gastroenterology and Hepatology, Renmin Hospital, Wuhan University, Wuhan, 430060, China
| | - Xiao-Jie Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China
| | - Xiao-Xin-Ran Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China
| | - Jiang-Feng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China.
| | - Yi-Ran Ni
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China.
| |
Collapse
|
13
|
Li K, Dai YJ, Zhang H, Zhang Z. YAP1 activates SLC2A1 transcription and augments the malignant behavior of colorectal cancer cells by activating the Wnt/β-catenin signaling pathway. Cell Div 2025; 20:8. [PMID: 40186232 PMCID: PMC11969700 DOI: 10.1186/s13008-025-00148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This paper examined the role of solute carrier family 2 member 1 (SLC2A1) in colorectal cancer (CRC) progression, focusing on its expression levels, functional implications, and regulatory mechanisms involving Yes-associated protein 1 (YAP1) and the Wnt signaling pathway. METHODS GEO datasets (GSE14297, GSE18462, GSE40367) were analyzed to identify genes linked to metastasis in CRC, and TCGA-COAD system was used to analyze the expression pattern and prognostic values of SLC2A1 in CRC. Functional studies were conducted using CRC cell lines (Caco-2 and SW480). Cell viability, migration and invasion, and apoptosis were examined using EdU assays, Transwell assays, and flow cytometry. YAP1's regulatory role on SLC2A1 was investigated using ChIP-qPCR and luciferase reporter assays. The Wnt/β-catenin agonist SKL2001 was used for functional rescue experiments. RESULTS SLC2A1 was upregulated in CRC cells, and its upregulation was associated with tumor metastasis and unfavorable outcomes according to bioinformatics. Knockdown of SLC2A1 resulted in reduced cell viability, decreased migration, and increased apoptosis in Caco-2 and SW480 cells. Additionally, YAP1 was identified as a transcriptional activator of SLC2A1. Knockdown of YAP1 decreased SLC2A1 expression and reduced expression of Wnt target genes, thus suppressing malignant behavior of tumor cells. However, further overexpression of SLC2A1 restored cell viability and migration in YAP1-deficient cells. The YAP1- SLC2A1 axis activated the Wnt/β-catenin by reducing GSK3β activity. CONCLUSION SLC2A1 is critical in CRC progression, with YAP1 serving as a key regulator of its expression and function. The YAP1-SLC2A1-Wnt axis represents a potential therapeutic target for CRC, providing insights into metabolic adaptations that support tumor growth and metastasis.
Collapse
Affiliation(s)
- Kunpeng Li
- Zhongda Hospital of Southeast University, No 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, PR China
| | - Ya-Jie Dai
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Haifeng Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Zhigang Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
14
|
Wang Z, Zhu P, Li H, Ye B, Luo Q, Cheng J, Cai Y. Sodium Hyaluronate-PDGF Repairs Cartilage and Subchondral Bone Microenvironment via HIF-1α-VEGF-Notch and SDF-1-CXCR4 Inhibition in Osteoarthritis. J Cell Mol Med 2025; 29:e70515. [PMID: 40159624 PMCID: PMC11955409 DOI: 10.1111/jcmm.70515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
Chronic degenerative changes in cartilage and subchondral bone that lead to instability of the cartilage microenvironment are essential for the development of osteoarthritis (OA) in the old. Synchronous repair of cartilage and subchondral bone may be a key strategy for OA treatment. PDGF-BB effectively promoted chondrocyte regeneration and angiogenesis. However, the mechanisms by which PDGF-BB affects subchondral bone and the delivery of PDGF-BB to the joint cavity need to be further explored. In this study, we used sodium hyaluronate to deliver PDGF-BB (SH-PDGF) to the joint space and aimed to determine the mechanisms of SH-PDGF in repairing cartilage and subchondral bone and stabilising the cartilage microenvironment. In this research, we determined the pharmacokinetics of PDGF-BB and SH-PDGF in cartilage. Moreover, we investigated the effects of PDGF-BB and SH-PDGF on cartilage and the subchondral bone microenvironment by identifying changes in the HIF-VEGF-Notch axis and SDF-1-CXCR4 axis in an OA rat model. The results showed that PDGF-BB increased cell viability, decreased HIF-1α levels, inhibited inflammation and improved matrix metabolism in osteoarthritic chondrocytes under hyperoxic or hypoxic conditions. We also found that PDGF-BB and SH-PDGF showed similar effects on repairing cartilage and subchondral bone simultaneously. However, SH-PDGF had some advantages over PDGF-BB in prolonging the injection interval and decreasing the injection time. These protective effects were mediated by the inhibition of both the HIF-1α-VEGF-Notch axis and the SDF-1-CXCR4 axis. The underlying mechanisms include the inhibition of HIF-1α-VEGF-Notch-mediated vessel invasion and SDF-1-CXCR4 axis-mediated crosstalk between cartilage and subchondral tissue.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Sports MedicineWuhan Fourth HospitalWuhanChina
- Hubei Provincial Sports Medicine CenterWuhanChina
| | - Pengfei Zhu
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of CardiovascularWuhan Fourth HospitalWuhanChina
| | - Hongmei Li
- Zibo First Hospital, Zibo Prevention and Treatment Hospital for Occupation DiseasesZiboChina
| | - Bo Ye
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| | - Qiong Luo
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| | - Jiangxia Cheng
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of AnesthesiologyWuhan Fourth HospitalWuhanChina
| | - Yu Cai
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| |
Collapse
|
15
|
Sencha LM, Karpova MA, Dobrynina OE, Balalaeva IV. Cell-type dependent effect of 3D collagen matrix on cancer cell resistance to suboptimal conditions: the case of serum deprivation, glucose starvation, and hypoxia. Tissue Cell 2025; 93:102719. [PMID: 39823703 DOI: 10.1016/j.tice.2024.102719] [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: 09/03/2024] [Revised: 12/13/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.
Collapse
Affiliation(s)
- Ludmila M Sencha
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria A Karpova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga E Dobrynina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| |
Collapse
|
16
|
Huang LX, Sun T, Sun J, Wu ZM, Zhao YB, Li MY, Huo QY, Ling C, Zhang BY, Chen C, Wang H. The Role of Endothelial Cell Glycolysis in Schwann Cells and Peripheral Nerve Injury Repair: A Novel and Important Research Area. Neurochem Res 2025; 50:121. [PMID: 40100469 DOI: 10.1007/s11064-025-04374-2] [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: 01/05/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Endothelial cell glycolysis plays a novel and significant role in Schwann cells and peripheral nerve injury repair, which represents an emerging and important area of research. Glycolysis in endothelial cells is a conserved and tightly regulated biological process that provides essential energy (ATP) and intermediates by ultimately converting glucose into lactate. This metabolic pathway is crucial for maintaining the normal function of endothelial cells. During peripheral nerve injury repair, endothelial cell glycolysis influences the function of Schwann cells and the efficiency of nerve regeneration. Beyond glycolysis, endothelial cells also secrete various factors, including growth factors and extracellular vesicles, which further modulate Schwann cell activity and contribute to the repair process. This review will summarize the role of endothelial cell glycolysis in Schwann cell function and peripheral nerve injury repair, aiming to provide new insights for the development of novel strategies for peripheral nerve injury treatment.
Collapse
Affiliation(s)
- Li-Xin Huang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Tao Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhi-Min Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Yi-Bo Zhao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ming-Yang Li
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Qing-Yi Huo
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Bao-Yu Zhang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
17
|
Rahman MA, Jalouli M, Bhajan SK, Al-Zharani M, Harrath AH. The Role of Hypoxia-Inducible Factor-1α (HIF-1α) in the Progression of Ovarian Cancer: Perspectives on Female Infertility. Cells 2025; 14:437. [PMID: 40136686 PMCID: PMC11941611 DOI: 10.3390/cells14060437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Hypoxia-Inducible Factor-1α (HIF-1α) is crucial in the progression of ovarian cancer, especially in influencing its tumor microenvironment and promoting pathogenic pathways that worsen female infertility. In hypoxic settings, HIF-1α is stabilized and activates the transcription of genes associated with angiogenesis, metabolic reprogramming, epithelial-to-mesenchymal transition, and therapeutic resistance. Angiogenesis and glycolytic reprogramming mediated by HIF-1 tumor proliferation, survival, and metastasis. Its dysfunction concurrently impairs ovarian homeostasis, undermining follicular growth, hormone synthesis, and the ovarian vascular network, consequently contributing to infertility. Moreover, HIF-1α induces persistent inflammation and oxidative stress, promoting an environment damaging to reproductive health. Due to its dual function in ovarian cancer growth and infertility, HIF-1α is a potential therapeutic target. Strategies including small molecule inhibitors and nanoparticle-mediated delivery of drugs possess the potential to reduce HIF-1α activity, hence reducing cancer progression while protecting fertility. This review seeks to clarify the molecular basis of HIF-1α in ovarian cancer and its effects on female infertility, providing insights into novel treatment approaches that target both controlling the disease and preserving fertility.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (M.J.); (M.A.-Z.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh;
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (M.J.); (M.A.-Z.)
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
McCaffrey EF, Delmastro AC, Fitzhugh I, Ranek JS, Douglas S, Peters JM, Fullaway CC, Bosse M, Liu CC, Gillen C, Greenwald NF, Anzick S, Martens C, Winfree S, Bai Y, Sowers C, Goldston M, Kong A, Boonrat P, Bigbee CL, Venugopalan R, Maiello P, Klein E, Rodgers MA, Scanga CA, Lin PL, Kirschner D, Fortune S, Bryson BD, Butler JR, Mattila JT, Flynn JL, Angelo M. The immunometabolic topography of tuberculosis granulomas governs cellular organization and bacterial control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638923. [PMID: 40027668 PMCID: PMC11870603 DOI: 10.1101/2025.02.18.638923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Despite being heavily infiltrated by immune cells, tuberculosis (TB) granulomas often subvert the host response to Mycobacterium tuberculosis (Mtb) infection and support bacterial persistence. We previously discovered that human TB granulomas are enriched for immunosuppressive factors typically associated with tumor-immune evasion, raising the intriguing possibility that they promote tolerance to infection. In this study, our goal was to identify the prime drivers for establishing this tolerogenic niche and to determine if the magnitude of this response correlates with bacterial persistence. To do this, we conducted a multimodal spatial analysis of 52 granulomas from 16 non-human primates (NHP) who were infected with low dose Mtb for 9-12 weeks. Notably, each granuloma's bacterial burden was individually quantified allowing us to directly ask how granuloma spatial structure and function relate to infection control. We found that a universal feature of TB granulomas was partitioning of the myeloid core into two distinct metabolic environments, one of which is hypoxic. This hypoxic environment associated with pathologic immune cell states, dysfunctional cellular organization of the granuloma, and a near-complete blockade of lymphocyte infiltration that would be required for a successful host response. The extent of these hypoxia-associated features correlated with worsened bacterial burden. We conclude that hypoxia governs immune cell state and organization within granulomas and is a potent driver of subverted immunity during TB.
Collapse
Affiliation(s)
- Erin F. McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
- Spatial Immunology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD
| | - Alea C. Delmastro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Isobel Fitzhugh
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Jolene S. Ranek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sarah Douglas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Joshua M. Peters
- Department of Biological Engineering, MIT, Cambridge, MA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
| | | | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Candace C. Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Craig Gillen
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Noah F. Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sarah Anzick
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Craig Martens
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Seth Winfree
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Yunhao Bai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Cameron Sowers
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Mako Goldston
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Potchara Boonrat
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Carolyn L. Bigbee
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Roopa Venugopalan
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Edwin Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Philana Ling Lin
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Pediatrics, Division of Infectious Disease, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Denise Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Sarah Fortune
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, MA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
| | - J. Russell Butler
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Joshua T. Mattila
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
19
|
Yang B, Liang H, Xu J, Liu Y, Ma S, Li Y, Wang C. Multi-drug sequential release systems: Construction and application for synergistic tumor treatment. Int J Pharm 2025; 670:125156. [PMID: 39746586 DOI: 10.1016/j.ijpharm.2024.125156] [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: 10/10/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
In tumor treatment, the sequence and timing of drug action have a large influence on therapeutic efficacy. Multi-drug sequential release systems (MDSRS) enable the sequential and/or on-demand release of multiple drugs following the single administration of a therapeutic agent. Several researchers have explored MDSRS, providing fresh strategies for synergistic cancer therapy. This review article first introduces the main characteristics of MDSRS. It then elaborates on the design principles of MDSRS. Subsequently, it summarizes the various structures of carriers used for constructing MDSRS, including core-shell structure, Layer-by-layer structure, Janus structure and hydrogel. Next, through specific examples, the article emphasizes the application of MDSRS in cancer treatment, focusing on their role in remodeling the tumor microenvironment (TME) and enhancing therapeutic effects through multiple mechanisms. Finally, the article discusses the current limitations and challenges of these systems and proposes potential future solutions. Overall, this review underscores the importance of the sequence and timing of drug therapy in cancer treatment, providing valuable theoretical and technical guidance for pharmaceutical research.
Collapse
Affiliation(s)
- Boyuan Yang
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Huijuan Liang
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Jiahao Xu
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Yanchi Liu
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Sha Ma
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Yuqiu Li
- School of Life Science and Technology, Kunming University of Science and Technology, China
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, China.
| |
Collapse
|
20
|
Wang Z, Shi Y, Gao B, Dang Z, Yang S, Chung CH, Yu X, Zhou X, Lin Z, Cheang LH, Tam MS, Wang H, Zheng X, Wu T. Development of a multi-functional naringin-loaded bioglass/carboxymethyl chitosan/silk fibroin porous scaffold for hemostasis and critical size bone regeneration. Int J Biol Macromol 2025; 290:138888. [PMID: 39701255 DOI: 10.1016/j.ijbiomac.2024.138888] [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: 07/30/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Persistent bleeding and limited repair capacity greatly threaten patients with bone destruction. Designing inorganic-organic biomimetic scaffolds with quick hemostasis and osteogenesis functions will solve this problem. A novel degradable and naringin (NG) loaded porous scaffold (SCB-N) based on APTES-modified bioactive glass (ABG), carboxymethyl chitosan and silk fibroin is developed. ABG and NG enhance the strength of the scaffolds. The scaffolds can release NG and bioactive ions (Ca2+ and Si4+), promoting the expression of osteogenesis (OCN, BMP-2), angiogenesis (VEGF), and neurogenesis (TB3, GFAP) genes in bone mesenchymal stem cells (BMSCs) and the related proteins (OCN, BMP-2, VEGF, GFAP). When implanting the scaffolds in rat cranial critical size defects, all scaffolds exhibit good compatibility, and SCB-N2 (with ABG and 1 mg/mL NG) group significantly promotes new bone regeneration and the formation of M2-type macrophages. Transcriptome sequencing results confirmed the osteogenic differentiation of BMSCs stimulated by SCB-N2 scaffolds is mainly regulated through MAPK and Wnt signaling pathways. Moreover, SCB-N2 group demonstrates quick hemostasis in vitro and in vivo due to the high adsorption ability and Ca ions release. The novel bionic scaffolds loaded with ion/traditional Chinese medicine monomer, possess the capabilities of hemostasis, neurovascularization, osteogenesis and immunomodulation, therefore exhibiting potential in bone repair.
Collapse
Affiliation(s)
- Zhaozhen Wang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China; Orthopedic and traumatology department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yiwan Shi
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China
| | - Botao Gao
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Zhaohui Dang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Shan Yang
- General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Chia-Hsuan Chung
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China
| | - Xiaolu Yu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China
| | - Xinting Zhou
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Zefeng Lin
- General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Lek Hang Cheang
- Department of Orthopedic Surgery, Centro Hospitalar Conde de Sao Januario, Macau
| | | | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
21
|
Zhou S, Sun J, Zhu W, Yang Z, Wang P, Zeng Y. Hypoxia studies in non‑small cell lung cancer: Pathogenesis and clinical implications (Review). Oncol Rep 2025; 53:29. [PMID: 39749693 PMCID: PMC11715622 DOI: 10.3892/or.2024.8862] [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: 10/11/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Non‑small cell lung cancer (NSCLC) is one of the most prevalent and lethal types of cancers worldwide and its high incidence and mortality rates pose a significant public health challenge. Despite significant advances in targeted therapy and immunotherapy, the overall prognosis of patients with NSCLC remains poor. Hypoxia is a critical driving factor in tumor progression, influencing the biological behavior of tumor cells through complex molecular mechanisms. The present review systematically examined the role of the hypoxic microenvironment in NSCLC, demonstrating its crucial role in promoting tumor cell growth, invasion and metastasis. Additionally, it has been previously reported that the hypoxic microenvironment enhances tumor cell resistance by activating hypoxia‑inducible factor and regulating exosome secretion. The hypoxic microenvironment also enables tumor cells to adapt to low oxygen and nutrient‑deficient conditions by enhancing metabolic reprogramming, such as through upregulating glycolysis. Further studies have shown that the hypoxic microenvironment facilitates immune escape by modulating tumor‑associated immune cells and suppressing the antitumor response of the immune system. Moreover, the hypoxic microenvironment increases tumor resistance to radiotherapy, chemotherapy and other types of targeted therapy through various pathways, significantly reducing the therapeutic efficacy of these treatments. Therefore, it could be suggested that early detection of cellular hypoxia and targeted therapy based on hypoxia may offer new therapeutic approaches for patients with NSCLC. The present review not only deepened the current understanding of the mechanisms of action and role of the hypoxic microenvironment in NSCLC but also provided a solid theoretical basis for the future development of precision treatments for patients with NSCLC.
Collapse
Affiliation(s)
- Sirui Zhou
- Department of Respiration, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Jiazheng Sun
- Department of Respiration, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Weijian Zhu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Zhiying Yang
- Department of Radiation Oncology, Minda Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Ping Wang
- Department of Respiration, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Yulan Zeng
- Department of Respiration, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| |
Collapse
|
22
|
Ai Z, Li D, Lan S, Zhang C. Nanomaterials exert biological effects by influencing the ubiquitin-proteasome system. Eur J Med Chem 2025; 282:116974. [PMID: 39556894 DOI: 10.1016/j.ejmech.2024.116974] [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: 09/03/2024] [Revised: 10/05/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024]
Abstract
The ubiquitin-proteasome system (UPS) is an important type of protein post-translational modification that affects the quantity and quality of various proteins and influences cellular processes such as the cell cycle, transcription, oxidative stress, and autophagy. Nanomaterials (NMs), which exhibit excellent physicochemical properties, can directly interact with the UPS and act as molecular-targeted drugs to induce changes in biological processes. This review provides an overview of the influence of NMs on the UPS of misfolded proteins and key proteins, which are related to cancer, neurodegenerative diseases and oxidative stress. This review also summarizes the role of modification processes involved in ubiquitination the biological effects of NMs and the mechanism of such effects of NMs through regulation of the UPS. This review deepens our understanding of the influence of NMs on the protein degradation process and provides new potential therapeutic targets for disease.
Collapse
Affiliation(s)
- Zhen Ai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Dan Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Shuquan Lan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
23
|
Zhang P, Zhang J, Ma C, Ma H, Jing L. 6-hydroxygenistein attenuates hypoxia-induced injury via activating Nrf2/HO-1 signaling pathway in PC12 cells. Sci Rep 2025; 15:875. [PMID: 39762378 PMCID: PMC11704347 DOI: 10.1038/s41598-025-85286-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
4',5,6,7-tetrahydoxyisoflavone (6-hydroxygenistein, 6-OHG) is a hydroxylated derivative of genistein with excellent antioxidant activity, but whether 6-OHG can protect hypoxia-induced damage is unclear. The objective of current study was to evaluate the protective effect and underling mechanism of 6-OHG against hypoxia-induced injury via network pharmacology and cellular experiments. 6-OHG-related and hypoxia injury-related targets were screened by public databases. The intersected targets were used for constructing PPI network and performing GO and KEGG functional analysis. We induced injury in PC12 cells under hypoxia conditions and observed the effects and molecular mechanisms of 6-OHG on cellular damage. Network pharmacological analysis predicted that 6-OHG delayed hypoxia injury by mitigating oxidative stress, inflammatory response and apoptosis. Cellular experiments suggested that 6-OHG treatment mitigated cell damage, enhanced cell viability, reduced ROS production and MDA level, increased SOD and CAT activities and elevated GSH level in PC12 cell exposed to hypoxia. Additionally, 6-OHG treatment reduced the TNF-α and IL-6 levels and elevated the IL-10 content, while downregulated the NF-κB and TNF-α expressions. 6-OHG also inhibited the caspase-3 and - 9 activation and the Bax and cleaved caspase-3 expressions, and elevated the Bcl-2 expression. Moreover, 6-OHG remarkably enhanced Nrf2 nuclear translocation and increased HO-1 expression. Molecular docking also proved the strong binding affinities of 6-OHG with Nrf2 and HO-1. Furthermore, ML385, a specific Nrf2 inhibitor, eliminated the beneficial effects of 6-OHG. In summary, 6-OHG can alleviate hypoxia-induced injury in PC12 cells through activating Nrf2/HO-1 signaling pathway and may be developed as candidate for preventing neuro-damage induced by hypoxia.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, NO.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, People's Republic of China
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support force of PLA, NO.333 Binhe South Road, Qilihe District, Lanzhou, 730050, Gansu, People's Republic of China
| | - Jie Zhang
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support force of PLA, NO.333 Binhe South Road, Qilihe District, Lanzhou, 730050, Gansu, People's Republic of China
| | - Chuan Ma
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support force of PLA, NO.333 Binhe South Road, Qilihe District, Lanzhou, 730050, Gansu, People's Republic of China
| | - Huiping Ma
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support force of PLA, NO.333 Binhe South Road, Qilihe District, Lanzhou, 730050, Gansu, People's Republic of China.
| | - Linlin Jing
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, NO.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, People's Republic of China.
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support force of PLA, NO.333 Binhe South Road, Qilihe District, Lanzhou, 730050, Gansu, People's Republic of China.
| |
Collapse
|
24
|
Tourn J, Crescence L, Bruzzese L, Panicot-Dubois L, Dubois C. Cellular and Molecular Mechanisms Leading to Air Travel-Induced Thrombosis. Circ Res 2025; 136:115-134. [PMID: 39745986 DOI: 10.1161/circresaha.124.325208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Venous thromboembolism, characterized by deep vein thrombosis and pulmonary embolism, is the third cardiovascular disease in the world. Deep vein thrombosis occurs when a blood clot forms in areas of impaired blood flow, and it is significantly affected by environmental factors. Local hypoxia, caused by venous stasis, plays a critical role in deep vein thrombosis under normal conditions, and this effect is intensified when the Po2 decreases, such as during air travel or high-altitude exposure. The lower oxygen levels and reduced pressure at high altitudes further contribute to deep vein thrombosis development. These conditions increase the pro-coagulant activity of neutrophils, platelets, and red blood cells, which interact on the surface of activated endothelial cells, promoting clot formation. Understanding the mechanisms involved in thrombus formation when Po2 is reduced, with or without pressure reduction, is crucial for preventing the development of venous thromboembolisms in such conditions and identifying innovative therapeutic targets. This literature review explores the mechanisms involved in thrombus formation related to high-altitude conditions and discusses the pro-coagulant consequences induced by environmental disturbances.
Collapse
Affiliation(s)
- Julie Tourn
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurie Bruzzese
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| |
Collapse
|
25
|
Wang L, Zhang J, Ye S, Lu F. LncRNA H19 improves mesenchymal characteristics of buffalo (Bubalus bubalis) bone marrow-derived mesenchymal stem cells under hypoxic conditions. Res Vet Sci 2025; 182:105461. [PMID: 39612735 DOI: 10.1016/j.rvsc.2024.105461] [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: 05/06/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024]
Abstract
As adult stem cells with various advantages, Bone marrow-derived mesenchymal stem cells (BMSCs) are valuable resources for veterinary treatment and animal reproduction. Previous studies have shown that hypoxia can induce epithelial-mesenchymal transition (EMT) and improve mesenchymal characteristics of BMSCs in vitro culture. However, the mechanism by which hypoxia improves the interstitial characteristics of buffalo BMSCs (bBMSCs) remains unclear. In this study, the effects of hypoxia on the mesenchymal characteristics of bBMSCs and the expression level of lncRNA H19 were examined, and then the effects of lncRNA H19 on maintaining the mesenchymal characteristics of bBMSCs under hypoxic culture conditions (5 % oxygen) as well as its mechanism also were explored, so as to further understand the molecular mechanism of mesenchymal characteristics maintenance of bBMSCs. The results showed that hypoxic culture conditions promoted EMT of bBMSCs, with lncRNA H19 expression up-regulated. When lncRNA H19 was knocked down in hypoxia, the expression level of Vimentin was down-regulated, the expression level of E-Cadherin was up-regulated, and EMT was inhibited. Meanwhile, the genes (p-PI3K and p-AKT1) involved in PI3K/AKT signaling pathway were inhibited by lncRNA H19 Knockdown. IGF-1 (10 ng/mL), an activator of PI3K/AKT signaling pathway, was added to rescued the inhibition of PI3K/AKT signaling pathway caused by lncRNA H19 Knockdown, with the effects of lncRNA H19 on EMT related genes also partially reversed. These findings not only provide theoretical guidance to elucidate the detailed regulation mechanism of hypoxia on mesenchymal nature maintenance of bBMSCs, but also provide positive support to further establish the stable in vitro culture system of bBMSCs.
Collapse
Affiliation(s)
- Lulu Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding and Diease Control, Guangxi University, Nanning 530005, China
| | - Jun Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, China
| | - Sheng Ye
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding and Diease Control, Guangxi University, Nanning 530005, China
| | - Fenghua Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding and Diease Control, Guangxi University, Nanning 530005, China.
| |
Collapse
|
26
|
Saber S, Nasr M, Yahya G, Elagamy HI, Abo Zaid MH, Sharaf H, Kira AY. Silk fibroin/gelatin electrospun nanofibrous dressing loaded with roxadustat accelerates wound healing in diabetic rats via HIF-1α stabilization. J Drug Deliv Sci Technol 2025; 103:106439. [DOI: 10.1016/j.jddst.2024.106439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
27
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
28
|
Li X, Li X, Qin J, Lei L, Guo H, Zheng X, Zeng X. Machine learning-derived peripheral blood transcriptomic biomarkers for early lung cancer diagnosis: Unveiling tumor-immune interaction mechanisms. Biofactors 2025; 51:e2129. [PMID: 39415336 DOI: 10.1002/biof.2129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
Lung cancer continues to be the leading cause of cancer-related mortality worldwide. Early detection and a comprehensive understanding of tumor-immune interactions are crucial for improving patient outcomes. This study aimed to develop a novel biomarker panel utilizing peripheral blood transcriptomics and machine learning algorithms for early lung cancer diagnosis, while simultaneously providing insights into tumor-immune crosstalk mechanisms. Leveraging a training cohort (GSE135304), we employed multiple machine learning algorithms to formulate a Lung Cancer Diagnostic Score (LCDS) based on peripheral blood transcriptomic features. The LCDS model's performance was evaluated using the area under the receiver operating characteristic (ROC) curve (AUC) in multiple validation cohorts (GSE42834, GSE157086, and an in-house dataset). Peripheral blood samples were obtained from 20 lung cancer patients and 10 healthy control subjects, representing an in-house cohort recruited at the Sixth People's Hospital of Chengdu. We employed advanced bioinformatics techniques to explore tumor-immune interactions through comprehensive immune infiltration and pathway enrichment analyses. Initial screening identified 844 differentially expressed genes, which were subsequently refined to 87 genes using the Boruta feature selection algorithm. The random forest (RF) algorithm demonstrated the highest accuracy in constructing the LCDS model, yielding a mean AUC of 0.938. Lower LCDS values were significantly associated with elevated immune scores and increased CD4+ and CD8+ T-cell infiltration, indicative of enhanced antitumor-immune responses. Higher LCDS scores correlated with activation of hypoxia, peroxisome proliferator-activated receptor (PPAR), and Toll-like receptor (TLR) signaling pathways, as well as reduced DNA damage repair pathway scores. Our study presents a novel, machine learning-derived peripheral blood transcriptomic biomarker panel with potential applications in early lung cancer diagnosis. The LCDS model not only demonstrates high accuracy in distinguishing lung cancer patients from healthy individuals but also offers valuable insights into tumor-immune interactions and underlying cancer biology. This approach may facilitate early lung cancer detection and contribute to a deeper understanding of the molecular and cellular mechanisms underlying tumor-immune crosstalk. Furthermore, our findings on the relationship between LCDS and immune infiltration patterns may have implications for future research on therapeutic strategies targeting the immune system in lung cancer.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xuebing Li
- Department of Respiratory and Critical Care Medicine, People's Hospital of Yaan, Yaan, Sichuan, China
| | - Jiangyue Qin
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Lei
- Department of Oncology, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hua Guo
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xi Zheng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuefeng Zeng
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Qu Y, Zeng A, Cheng Y, Li S. Natural killer cell memory: challenges and opportunities for cancer immunotherapy. Cancer Biol Ther 2024; 25:2376410. [PMID: 38987282 PMCID: PMC11238922 DOI: 10.1080/15384047.2024.2376410] [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: 02/05/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Substantial advancements have been made in recent years in comprehending immune memory, which enhances the secondary response through prior infections. The ability of vertebrate T and B lymphocytes to exhibit classic recall responses has long been regarded as a distinguishing characteristic. However, natural killer (NK) cells have been found to acquire immunological memory in a manner akin to T and B cells. The fundamental principles derived from the investigation of NK cell memory offer novel insights into innate immunity and have the potential to pave the way for innovative strategies to enhance therapeutic interventions against multiple diseases including cancer. Here, we reviewed the fundamental characteristics, memory development and regulatory mechanism of NK cell memory. Moreover, we will conduct a comprehensive evaluation of the accomplishments, obstacles, and future direction pertaining to the utilization of NK cell memory in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Yuhua Qu
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anhui Zeng
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulu Cheng
- Department of Disinfection Supply Center, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengchun Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
30
|
Seo SH, Lee JH, Choi EK, Rho SB, Yoon K. C/EBPβ Regulates HIF-1α-Driven Invasion of Non-Small-Cell Lung Cancer Cells. Biomolecules 2024; 15:36. [PMID: 39858431 PMCID: PMC11764306 DOI: 10.3390/biom15010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Metastatic cancer accounts for most cancer-related deaths, and identifying specific molecular targets that contribute to metastatic progression is crucial for the development of effective treatments. Hypoxia, a feature of solid tumors, plays a role in cancer progression by inducing resistance to therapy and accelerating metastasis. Here, we report that CCAAT/enhancer-binding protein beta (C/EBPβ) transcriptionally regulates hypoxia-inducible factor 1 subunit alpha (HIF1A) and thus promotes migration and invasion of non-small-cell lung cancer (NSCLC) cells under hypoxic conditions. Our results show that knockdown or forced expression of C/EBPβ was correlated with HIF-1α expression and that C/EBPβ directly bound to the promoter region of HIF1A. Silencing HIF1A inhibited the enhanced migration and invasion induced by C/EBPβ overexpression in NSCLC cells under hypoxia. Expression of the HIF-1α target gene, SLC2A1, was also altered in a C/EBPβ-dependent manner, and knockdown of SLC2A1 reduced migration and invasion enhanced by C/EBPβ overexpression. These results indicate that C/EBPβ is a critical regulator for the invasion of NSCLC cells in the hypoxic tumor microenvironment. Collectively, the C/EBPβ-HIF-1α-GLUT1 axis represents a potential therapeutic target for preventing metastatic progression of NSCLC and improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Kyungsil Yoon
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.H.S.); (J.H.L.); (S.B.R.)
| |
Collapse
|
31
|
Yang JL, Yang J, Fang RF, Sai WL, Yao DF, Yao M. Hypoxia upregulates hepatic angiopoietin-2 transcription to promote the progression of hepatocellular carcinoma. World J Hepatol 2024; 16:1480-1492. [DOI: 10.4254/wjh.v16.i12.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Angiopoietin-2 (Ang-2) level is related to hepatocellular carcinoma (HCC) progression. However, the dynamic expression and regulatory mechanism of Ang-2 remain unclear.
AIM To investigate Ang-2 levels in chronic liver diseases and validate early monitoring value with a dynamic model in hepatocarcinogenesis.
METHODS Sprague-Dawley rats in hepatocarcinogenesis were induced with diet 2-fluorenylacet-amide, and grouped based on liver histopathology by hematoxylin and eosin staining. Differently expressed genes or Ang-2 mRNA in livers were analyzed by whole-genome microarray. Ang-2 levels in chronic liver diseases were detected by an enzyme-linked immunosorbent assay.
RESULTS Clinical observation reveled that the circulating levels of Ang-2 and hypoxia-inducible factor-1α (HIF-1α) in patients with chronic liver diseases were progressively increased from benign to HCC (P < 0.001). Dynamic model validated that the up-regulated Ang-2 in liver and blood was positively correlated with HIF-1α in hepatocarcinogenesis (P < 0.001). Mechanistically, Ang-2 was regulated by HIF-1α. When specific HIF-1α- microRNAs transfected into HCC cells, the cell proliferation significantly inhibited, HIF-1α and Ang-2 down-regulated, and also affected epithelial-mesenchymal transition via increasing E-cadherin to block cell invasion or migration with reducing of snail, twist and vimentin.
CONCLUSION Hypoxia-induced Ang-2 up-regulating expression might serve as a sensitive early monitoring biomarker for hepatocarcinogenesis or HCC metastasis.
Collapse
Affiliation(s)
- Jun-Ling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jie Yang
- Department of Biology, Life Science School of Nantong University, Nantong 226009, Jiangsu Province, China
| | - Rong-Fei Fang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wen-Li Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Min Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
32
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [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: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
33
|
Yan T, Shi J. Angiogenesis and EMT regulators in the tumor microenvironment in lung cancer and immunotherapy. Front Immunol 2024; 15:1509195. [PMID: 39737184 PMCID: PMC11682976 DOI: 10.3389/fimmu.2024.1509195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Lung cancer remains the primary cause of cancer-related mortality, with factors such as postoperative tumor recurrence, metastasis, and therapeutic drug resistance exacerbating patient outcomes. Immunotherapy has emerged as a transformative approach, challenging conventional treatment paradigms for lung cancer. Consequently, advancing research in lung cancer immunotherapy is imperative. Recent studies indicate that numerous regulators within the tumor microenvironment (TME) drive tumor angiogenesis and epithelial-mesenchymal transition (EMT); these processes are interdependent, reciprocal, and collectively contribute to tumor progression. Tumor angiogenesis not only supplies adequate oxygen and nutrients for cellular proliferation but also establishes pathways facilitating tumor metastasis and creating hypoxic regions that foster drug resistance. Concurrently, EMT enhances metastatic potential and reinforces drug-resistance genes within tumor cells, creating a reciprocal relationship with angiogenesis. This interplay ultimately results in tumor invasion, metastasis, and therapeutic resistance. This paper reviews key regulators of angiogenesis and EMT, examining their impact on lung cancer immunotherapy and progression, and investigates whether newly identified regulators could influence lung cancer treatment, thus offering valuable insights for developing future therapeutic strategies.
Collapse
Affiliation(s)
- Taotao Yan
- Medical School of Nantong University, Nantong University, Nantong, China
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
34
|
Kim J, Eo EY, Kim B, Lee H, Kim J, Koo BK, Kim HJ, Cho S, Kim J, Cho YJ. Transcriptomic Analysis of Air-Liquid Interface Culture in Human Lung Organoids Reveals Regulators of Epithelial Differentiation. Cells 2024; 13:1991. [PMID: 39682739 PMCID: PMC11639892 DOI: 10.3390/cells13231991] [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: 10/07/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
To develop in vitro respiratory models, it is crucial to identify the factors involved in epithelial cell differentiation. In this study, we comprehensively analyzed the effects of air-liquid interface (ALI) culture on epithelial cell differentiation using single-cell RNA sequencing (scRNA-seq). ALI culture induced a pronounced shift in cell composition, marked by a fivefold increase in ciliated cells and a reduction of more than half in basal cells. Transcriptional signatures associated with epithelial cell differentiation, analyzed using iPathwayGuide software, revealed the downregulation of VEGFA and upregulation of CDKN1A as key signals for epithelial differentiation. Our findings highlight the efficacy of the ALI culture for replicating the human lung airway epithelium and provide valuable insights into the crucial factors that influence human ciliated cell differentiation.
Collapse
Affiliation(s)
- Jieun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (J.K.); (E.-Y.E.); (B.K.); (H.-J.K.)
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
| | - Eun-Young Eo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (J.K.); (E.-Y.E.); (B.K.); (H.-J.K.)
| | - Bokyong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (J.K.); (E.-Y.E.); (B.K.); (H.-J.K.)
| | - Heetak Lee
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea; (H.L.); (J.K.); (B.-K.K.)
| | - Jihoon Kim
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea; (H.L.); (J.K.); (B.-K.K.)
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea; (H.L.); (J.K.); (B.-K.K.)
| | - Hyung-Jun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (J.K.); (E.-Y.E.); (B.K.); (H.-J.K.)
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Jinho Kim
- Department of Genomic Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (J.K.); (E.-Y.E.); (B.K.); (H.-J.K.)
| |
Collapse
|
35
|
Liu WJ, Wang L, Sun FL, Zhou FM, Zhang RK, Liu J, Zhao M, Wang LH, Qin YR, Zhao YQ, Qiu JG, Jiang BH. Hexavalent chromium induced metabolic reprogramming, carcinogenesis and tumor progression through PDK1 upregulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117341. [PMID: 39550876 DOI: 10.1016/j.ecoenv.2024.117341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Lung cancer is the leading factor of cancer-related death in the worldwide. Hexavalent chromium [Cr(VI)] is a potential carcinogen for inducing lung cancers. To understand new mechanism of Cr(VI)-induced tumorigenesis and cancer development, we identified that PDK1 expression levels were greatly increased in chromium-transformed cells (Cr-T) compared to the parental BEAS-2B (B2B) cells by proteomic profiling and Western blotting; PDK1 levels were also induced in lung cancer cell lines and in lung samples of mice exposed to Cr(VI). Cr(VI) increased Warburg effect, cell migration, proliferation and colony formation through PDK1 upregulation. To identify the mechanism of PDK1 induction, we performed miRNA-seq analysis of Cr-T and B2B cells, and found miR-493 levels was significantly suppressed by Cr(VI). PDK1 was induced by miR-493 suppression, and was a direct target of miR-493. Interestingly, we also found HIF-1α was directly targeting by miR-493 and was induced by miR-493 downregulation. HIF-1α expression levels were upregulated in lung samples of mice with Cr(VI)-exposure. PDK1 was induced by HIF-1α, showing miR-493 suppression can directly induce PDK1 as well as through HIF-1α induction. MiR-493 overexpression was sufficient to suppress tumor growth, PDK1 and HIF-1α expression in vivo. We also showed that levels of miR-493 suppression, HIF-1α and PDK1 elevations were strongly correlated with poor prognosis of lung cancer subjects. These results demonstrate both HIF-1α and PDK1 expression are induced by Cr(VI)-mediated miR-493 suppression, and MiR-493/HIF-1α/PDK1 axis is a new pathway in Cr(VI)-inducing carcinogenesis and tumor growth.
Collapse
Affiliation(s)
- Wen-Jing Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lin Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Fan-Li Sun
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Feng-Mei Zhou
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Rui-Ke Zhang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Jie Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Min Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Li-Hong Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yan-Ru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yan-Qiu Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jian-Ge Qiu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Bing-Hua Jiang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
36
|
Hu Y, Fan Q, Qiao B, Xu O, Lv B, Han N, Zhang X. Alleviatory Role of Panax Notoginseng Saponins in Modulating Inflammation and Pulmonary Vascular Remodeling in Chronic Obstructive Pulmonary Disease: mechanisms and Implications. COPD 2024; 21:2329282. [PMID: 38622983 DOI: 10.1080/15412555.2024.2329282] [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: 10/17/2023] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
COPD is an inflammatory lung disease that limits airflow and remodels the pulmonary vascular system. This study delves into the therapeutic potential and mechanistic underpinnings of Panax notoginseng Saponins (PNS) in alleviating inflammation and pulmonary vascular remodeling in a COPD rat model. Symmap and ETCM databases provided Panax notoginseng-related target genes, and the CTD and DisGeNET databases provided COPD-related genes. Intersection genes were subjected to protein-protein interaction analysis and pathway enrichment to identify downstream pathways. A COPD rat model was established, with groups receiving varying doses of PNS and a Roxithromycin control. The pathological changes in lung tissue and vasculature were examined using histological staining, while molecular alterations were explored through ELISA, RT-PCR, and Western blot. Network pharmacology research suggested PNS may affect the TLR4/NF-κB pathway linked to COPD development. The study revealed that, in contrast to the control group, the COPD model exhibited a significant increase in inflammatory markers and pathway components such as TLR4, NF-κB, HIF-1α, VEGF, ICAM-1, SELE mRNA, and serum TNF-α, IL-8, and IL-1β. Treatment with PNS notably decreased these markers and mitigated inflammation around the bronchi and vessels. Taken together, the study underscores the potential of PNS in reducing lung inflammation and vascular remodeling in COPD rats, primarily via modulation of the TLR4/NF-κB/HIF-1α/VEGF pathway. This research offers valuable insights for developing new therapeutic strategies for managing and preventing COPD.
Collapse
Affiliation(s)
- Yanan Hu
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
- Heze Hospital of Traditional Chinese Medicine, Heze, P. R. China
| | - Qiuyang Fan
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
| | - Bo Qiao
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Ou Xu
- Fuwai Yunnan Cardiovascular Hospital, Kunming, P. R. China
| | - Bijun Lv
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
| | - Niping Han
- Molecular Biology for Sinomedicine, Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, P. R. China
| | - Xiaomei Zhang
- Molecular Biology for Sinomedicine, Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, P. R. China
| |
Collapse
|
37
|
Feizi N, Mohamadzadeh-Nabiei M, Vahedi H, Farabi Maleki S, Jafarizadeh A. Therapeutic role of erythropoietin in methanol induced optic neuropathy: a systematic review. Daru 2024; 33:2. [PMID: 39613913 PMCID: PMC11607285 DOI: 10.1007/s40199-024-00551-w] [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: 01/07/2024] [Accepted: 11/06/2024] [Indexed: 12/01/2024] Open
Abstract
PURPOSE Despite various therapeutic attempts, an approved treatment for Methanol-induced optic neuropathy (MION), a sight-threatening disorder, is still lacking. Erythropoietin known as an erythropoietic cytokine, possesses various non-hematopoietic properties that make it a candidate for MION treatment. This systematic review aims to assess the potential therapeutic role of erythropoietin in MION. METHOD We systematically searched English and Persian databases including PubMed, Scopus, Embase, Web of Science, and Scientific Information Database (SID) as of July 2024. Two independent authors screened the articles based on their titles, abstracts, and full texts to finalize the included articles in this study. The selected articles underwent quality assessments via the Joanna Briggs Institute (JBI) checklists. RESULTS Out of 139 studies identified in the databases, 11 were finally included in the analysis. These studies encompassed 212 participants, with 192 receiving erythropoietin treatment. Visual acuity (VA) improved in 184 patients, with improvements ranging from no light perception to full vision recovery, or minor enhancements such as an improvement from 1.75 ± 0.72 to 1.32 ± 0.79 LogMAR. Only 8 patients showed no change or experienced deterioration. Additionally, 21 cases exhibited a reduction in retinal nerve fiber layer thickness, with one showing a reduction towards the normal range. CONCLUSION This review highlights erythropoietin's positive impact on VA in patients with MION. However, simultaneous use of erythropoietin and corticosteroids in studies without control groups complicates evaluating erythropoietin's independent efficacy. Future research should involve large, controlled trials to clarify erythropoietin's role and establish it as a standard treatment. PROSPERO REGISTRATION NUMBER CRD42023485772.
Collapse
Affiliation(s)
- Neda Feizi
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Mohamadzadeh-Nabiei
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Vahedi
- Nikookari Eye Center, Tabriz University of Medical Sciences, Tabriz, 5166614766, Iran
| | - Shadi Farabi Maleki
- Nikookari Eye Center, Tabriz University of Medical Sciences, Tabriz, 5166614766, Iran
| | - Ali Jafarizadeh
- Nikookari Eye Center, Tabriz University of Medical Sciences, Tabriz, 5166614766, Iran.
| |
Collapse
|
38
|
Bou-Gharios J, Noël G, Burckel H. The neglected burden of chronic hypoxia on the resistance of glioblastoma multiforme to first-line therapies. BMC Biol 2024; 22:278. [PMID: 39609830 PMCID: PMC11603919 DOI: 10.1186/s12915-024-02075-w] [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: 06/06/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common adult primary brain tumor. The standard of care involves maximal surgery followed by radiotherapy and concomitant chemotherapy with temozolomide (TMZ), in addition to adjuvant TMZ. However, the recurrence rate of GBM within 1-2 years post-diagnosis is still elevated and has been attributed to the accumulation of multiple factors including the heterogeneity of GBM, genomic instability, angiogenesis, and chronic tumor hypoxia. Tumor hypoxia activates downstream signaling pathways involved in the adaptation of GBM to the newly oxygen-deprived environment, thereby contributing to the resistance and recurrence phenomena, despite the multimodal therapeutic approach used to eradicate the tumor. Therefore, in this review, we will focus on the development and implication of chronic or limited-diffusion hypoxia in tumor persistence through genetic and epigenetic modifications. Then, we will detail the hypoxia-induced activation of vital biological pathways and mechanisms that contribute to GBM resistance. Finally, we will discuss a proteomics-based approach to encourage the implication of personalized GBM treatments based on a hypoxia signature.
Collapse
Affiliation(s)
- Jolie Bou-Gharios
- Institut de Cancérologie Strasbourg Europe (ICANS), Radiobiology Laboratory, 3 Rue de La Porte de L'Hôpital, Strasbourg, 67000, France
- Laboratory of Engineering, Informatics and Imaging (ICube), UMR 7357, Integrative Multimodal Imaging in Healthcare (IMIS), University of Strasbourg, 4 Rue Kirschleger, Strasbourg, 67000, France
| | - Georges Noël
- Institut de Cancérologie Strasbourg Europe (ICANS), Radiobiology Laboratory, 3 Rue de La Porte de L'Hôpital, Strasbourg, 67000, France
- Laboratory of Engineering, Informatics and Imaging (ICube), UMR 7357, Integrative Multimodal Imaging in Healthcare (IMIS), University of Strasbourg, 4 Rue Kirschleger, Strasbourg, 67000, France
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, UNICANCER, 17 Rue Albert Calmette, Strasbourg, 67200, France
| | - Hélène Burckel
- Institut de Cancérologie Strasbourg Europe (ICANS), Radiobiology Laboratory, 3 Rue de La Porte de L'Hôpital, Strasbourg, 67000, France.
- Laboratory of Engineering, Informatics and Imaging (ICube), UMR 7357, Integrative Multimodal Imaging in Healthcare (IMIS), University of Strasbourg, 4 Rue Kirschleger, Strasbourg, 67000, France.
| |
Collapse
|
39
|
Zeng Q, Lv C, Qi L, Wang Y, Hao S, Li G, Sun H, Du L, Li J, Wang C, Zhang Y, Wang X, Ma R, Wang T, Li Q. Sodium selenite inhibits cervical cancer progression via ROS-mediated suppression of glucose metabolic reprogramming. Life Sci 2024; 357:123109. [PMID: 39384146 DOI: 10.1016/j.lfs.2024.123109] [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: 07/25/2024] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
AIMS This study aims to explore the inhibitory effect of selenium on cervical cancer through suppression of glucose metabolic reprogramming and its underlying mechanisms. METHODS Sodium selenite (SS) treated HeLa and SiHa cells were assessed for proliferation using the CCK-8 assay and immunofluorescence. DNA synthesis was measured with the EdU assay. A nude mouse xenograft model evaluated SS's anti-cervical cancer effects. Reactive oxygen species (ROS) and mitochondrial membrane potential were measured using flow cytometry, DCFH-DA, and JC-1 probes, respectively. Apoptosis was detected via Annexin V/PI staining and Western blot. Glucose uptake, lactate production, and ATP generation were determined using 2-NBDG probes and assay kits. The mRNA and protein levels of glycolysis-related genes HK2, GLUT1, and PDK1 were measured using RT-qPCR and Western blot. KEY FINDINGS SS inhibited HeLa and SiHa cells viability in a dose- and time-dependent manner. Intraperitoneal injection of SS in nude mice significantly inhibited HeLa cell xenograft growth without evident hepatotoxicity or nephrotoxicity. SS inhibited glucose metabolic reprogramming in cancer cells primarily via ROS-mediated AKT/mTOR/HIF-1α pathway inhibition. Pretreatment with N-acetylcysteine (NAC) or MHY1485 (an mTOR activator) partially reversed the inhibitory effects of SS on glucose metabolic reprogramming, cell proliferation, and migration, as well as its pro-apoptotic effects. SIGNIFICANCE SS exhibited anti-cervical cancer effects, likely through the induction of ROS generation and inhibition of glucose metabolic reprogramming in cervical cancer cells, thereby inhibiting cell proliferation and promoting apoptosis. These findings provide new insights into understanding the molecular mechanisms underlying SS for potential new drug development for cervical cancer.
Collapse
Affiliation(s)
- Qingyu Zeng
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| | - Cunqi Lv
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Lei Qi
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; School of Public Health, Qiqihar Medical University, Qiqihar 161003, Heilongjiang, China
| | - Yuanyuan Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Shuxiu Hao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Guijin Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Huixin Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Linlin Du
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Jiacheng Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Cheng Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Yu Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Xinshu Wang
- Nanchang University Queen Mary School, Nanchang 330000, China
| | - Rong Ma
- Department of Gynecological Oncoology, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| | - Tong Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| | - Qi Li
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| |
Collapse
|
40
|
Chen Y, Chen Y, Wang Z, Yang L, Zhang Y, Zhang Z, Jia L. Iron-based MOF with Catalase-like activity improves the synergistic therapeutic effect of PDT/ferroptosis/starvation therapy by reversing the tumor hypoxic microenvironment. J Nanobiotechnology 2024; 22:705. [PMID: 39543666 PMCID: PMC11562077 DOI: 10.1186/s12951-024-02921-7] [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: 09/11/2023] [Accepted: 10/10/2024] [Indexed: 11/17/2024] Open
Abstract
Reversing the hypoxic microenvironment of tumors is an important method to enhance the synergistic effect of tumor treatment. In this work, we developed the nanoparticles called Ce6@HGMOF, which consists of a photosensitizer (Ce6), glucose oxidase (GOX), chemotherapy drugs (HCPT) and an iron-based metal-organic framework (MOF). Ce6@HGMOF can consume glucose in tumor cells through "starvation therapy", cut off their nutrition source, and produce gluconic acid and hydrogen peroxide (H2O2). Utilizing this feature, Ce6@HGMOF can produce oxygen through catalase-like catalytic activity, thereby reversing the hypoxic microenvironment of tumors. This strategy of changing the hypoxic environment can help to slow down the growth of tumor blood vessels and improve the drug-resistant microenvironment to some extent. Meanwhile, increasing the supply of oxygen can enhance the effect of photodynamic therapy (PDT) and enhance the oxidative stress damage caused by reactive oxygen species (ROS) in tumor cells. On the other hand, cancer cells usually produce higher levels of glutathione (GSH) to adapt to high oxidative stress and protect themselves. The Ce6@HGMOF we designed can also consume GSH and induce ferroptosis of tumor cells through Fenton reaction with H2O2, while enhancing the effect of PDT. This innovative synergistic strategy, the combination of PDT/ferroptosis /starvation therapy, can complement each other and enhance each other. It has great potential as a powerful new anti-tumor paradigm in the future.
Collapse
Affiliation(s)
- Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenzhi Wang
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lian Yang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
41
|
Chen C, Qin S, Song X, Wen J, Huang W, Sheng Z, Li X, Cao Y. PI3K p85α/HIF-1α accelerates the development of pulmonary arterial hypertension by regulating fatty acid uptake and mitophagy. Mol Med 2024; 30:208. [PMID: 39528930 PMCID: PMC11552344 DOI: 10.1186/s10020-024-00975-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by lipid accumulation and mitochondrial dysfunction. This study was designed to investigate the effects of hypoxia-inducible factor-1α (HIF-1α) on fatty acid uptake and mitophagy in PAH. METHODS Peripheral blood samples were obtained from PAH patients. Human pulmonary arterial smooth muscle cells and rat cardiac myoblasts H9c2 were subjected to hypoxia treatment. Male Sprague-Dawley rats were treated with monocrotaline (MCT). Right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), pulmonary artery remodeling, and lipid accumulation were measured. Cell proliferation and ROS accumulation were assessed. Mitochondrial damage and autophagosome formation were observed. Co-immunoprecipitation was performed to verify the interaction between HIF-1α and CD36/PI3K p85α. RESULTS HIF-1α, CD36, Parkin, and PINK1 were upregulated in PAH samples. HIF-1α knockdown or PI3K p85α knockdown restricted the expression of HIF-1α, PI3K p85α, Parkin, PINK1, and CD36, inhibited hPASMC proliferation, promoted H9c2 cell proliferation, reduced ROS accumulation, and suppressed mitophagy. CD36 knockdown showed opposite effects to HIF-1α knockdown, which were reversed by palmitic acid. The HIF-1α activator dimethyloxalylglycine reversed the inhibitory effect of Parkin knockdown on mitophagy. In MCT-induced rats, the HIF-1α antagonist 2-methoxyestradiol (2ME) reduced RVSP, RVHI, pulmonary artery remodeling, lipid accumulation, and mitophagy. Recombinant CD36 abolished the therapeutic effect of 2ME but inhibited mitophagy. Activation of Parkin/PINK1 by salidroside (Sal) promoted mitophagy to ameliorate the pathological features of PAH-like rats, and 2ME further enhanced the therapeutic outcome of Sal. CONCLUSION PI3K p85α/HIF-1α induced CD36-mediated fatty acid uptake and Parkin/PINK1-dependent mitophagy to accelerate the progression of experimental PAH.
Collapse
Affiliation(s)
- Chenyang Chen
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Sirun Qin
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiaohua Song
- Department of Pediatrics, The 921, Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Changsha, 410011, China
| | - Juan Wen
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Wei Huang
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Zhe Sheng
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiaogang Li
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yu Cao
- Cardiovascular Department, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| |
Collapse
|
42
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [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: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Tan W, Chen G, Ci Q, Deng Z, Gu R, Yang D, Dai F, Liu H, Cheng Y. Elevated ITGA3 expression serves as a novel prognostic biomarker and regulates tumor progression in cervical cancer. Sci Rep 2024; 14:27063. [PMID: 39511266 PMCID: PMC11543847 DOI: 10.1038/s41598-024-75770-x] [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: 04/05/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Patients with advanced and recurrent cervical cancer often lack satisfactory treatment outcomes. Thus, it is necessary to seek reliable biomarkers that provide the ability to identify the disease at an early stage and predict the patient prognosis, providing new strategies for the treatment of cervical cancer. The sequencing data of ITGA3 were retrieved from public datasets. Immune infiltration and sensitivity of potential immunotherapy and chemotherapy have been analyzed between two subgroups. Functional analysis was applied to excavate the related pathways of ITGA3 in cervical cancer. Furthermore, the impact of ITGA3 in tumor progression has been verified in vitro. The results revealed that the level of ITGA3 was upregulated in cervical cancer, and was positively correlated with worse prognosis. The tumor microenvironment of patients in the high-risk group was immunosuppressed. Patients in high-risk group may not benefit from immunotherapy, but be may be sensitive to several chemotherapy drugs. Notably, the angiogenesis, epithelial mesenchymal transition, and PI3K pathway were increased in high-risk group. Collectively, ITGA3 is a marker of poor prognosis and promotes tumor progression by regulating PI3K/AKT pathway in cervical cancer. Our results provide new insights for potential molecular targeted therapy and prognostic prediction of cervical cancer.
Collapse
Affiliation(s)
- Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Gantao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qinyu Ci
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Ran Gu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
44
|
Liu X, Wang J, Shen K, Jin W. p53/HIF-1α regulates neuronal aging and autophagy in spinal cord ischemia/reperfusion injury. Mech Ageing Dev 2024; 222:112000. [PMID: 39515667 DOI: 10.1016/j.mad.2024.112000] [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: 04/22/2024] [Revised: 09/11/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
OBJECTION Spinal cord injury (SCI)-induced hindlimb dysfunction affects the physical and mental health of patients. There is growing evidence suggesting that the recovery capacity of elderly SCI patients is poorer than that of young individuals. However, the specific molecular mechanisms remain unclear. METHODS RNA expression profiles of SCI samples were collected from the GEO database, and key genes involved in the progression of SCI were identified by the limma package in R software. A diagnostic model based on SCIDEG was constructed using LASSO regression analysis. Subsequently, correlation analysis was conducted to identify biological pathways influenced by the key genes. Furthermore, SCI animal models were established in different age groups to examine the expression of relevant genes and verify the molecular mechanism of p53/HIF-1α axis. RESULTS We initially identified 34 ischemia-hypoxia-related genes potentially involved in the progression of SCI. Subsequently, we constructed a diagnostic model based on SCIDEGs using LASSO regression analysis. This model highlighted 9 key genes (TP53, SFTPA1, MASP2, KRT14, IL9, HIF1A, HGFAC, FUT7, and ALPP), which demonstrated high diagnostic accuracy in both the training set (AUC=1) and the validation set (AUC=0.855). Further cross-analysis with ischemia-reperfusion-related datasets confirmed the involvement of HIF1A and TP53. We also observed significant enrichment of HIF1A in organoids composed of mature neurons, which induced neuronal damage. In subsequent spinal cord injury animal models of different age groups, we found that HIF-1α expression was downregulated in the spinal cord tissues of elderly animals. Additionally, we discovered that TP53 activation induces cellular senescence in aging neurons and suppresses HIF-1α expression and autophagy levels within these cells. CONCLUSION In summary, our study suggests that the p53/HIF-1α signaling pathway plays a critical role in regulating neuronal aging and autophagy in the pathogenesis of SCI. Importantly, HIF-1α may represent a promising therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Xingzhen Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China
| | - Jia Wang
- Department of Pathology, Shanghai Xin Hua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, China
| | - Kangping Shen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China.
| | - Wenjie Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China.
| |
Collapse
|
45
|
Khattak S, Ullah I, Sohail M, Akbar MU, Rauf MA, Ullah S, Shen J, Xu H. Endogenous/exogenous stimuli‐responsive smart hydrogels for diabetic wound healing. AGGREGATE 2024. [DOI: 10.1002/agt2.688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
AbstractDiabetes significantly impairs the body's wound‐healing capabilities, leading to chronic, infection‐prone wounds. These wounds are characterized by hyperglycemia, inflammation, hypoxia, variable pH levels, increased matrix metalloproteinase activity, oxidative stress, and bacterial colonization. These complex conditions complicate effective wound management, prompting the development of advanced diabetic wound care strategies that exploit specific wound characteristics such as acidic pH, high glucose levels, and oxidative stress to trigger controlled drug release, thereby enhancing the therapeutic effects of the dressings. Among the solutions, hydrogels emerge as promising due to their stimuli‐responsive nature, making them highly effective for managing these wounds. The latest advancements in mono/multi‐stimuli‐responsive smart hydrogels showcase their superiority and potential as healthcare materials, as highlighted by relevant case studies. However, traditional wound dressings fall short of meeting the nuanced needs of these wounds, such as adjustable adhesion, easy removal, real‐time wound status monitoring, and dynamic drug release adjustment according to the wound's specific conditions. Responsive hydrogels represent a significant leap forward as advanced dressings proficient in sensing and responding to the wound environment, offering a more targeted approach to diabetic wound treatment. This review highlights recent advancements in smart hydrogels for wound dressing, monitoring, and drug delivery, emphasizing their role in improving diabetic wound healing. It addresses ongoing challenges and future directions, aiming to guide their clinical adoption.
Collapse
Affiliation(s)
- Saadullah Khattak
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Ihsan Ullah
- Zhejiang Engineering Research Center for Tissue Repair Materials Wenzhou Institute University of Chinese Academy of Sciences Wenzhou China
| | - Mohammad Sohail
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Muhammad Usman Akbar
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou China
| | - Mohd Ahmar Rauf
- Department of Internal Medicine, Heme Oncology Unit, University of Michigan Ann Arbor Michigan USA
| | - Salim Ullah
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry Eye Hospital Wenzhou Medical University Wenzhou China
- Wenzhou Institute University of Chinese Academy of Sciences Wenzhou China
| | - Hong‐Tao Xu
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| |
Collapse
|
46
|
Zhu B, Cheng L, Huang B, Liu R, Ren B. Central role of hypoxia-inducible factor-1α in metabolic reprogramming of cancer cells: A review. Medicine (Baltimore) 2024; 103:e40273. [PMID: 39496001 PMCID: PMC11537650 DOI: 10.1097/md.0000000000040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/09/2024] [Indexed: 11/06/2024] Open
Abstract
Metabolic reprogramming is one of the characteristics of tumor cell metabolism. In tumor cells, there are multiple metabolic enzymes and membrane proteins to regulate metabolic reprogramming, and hypoxia inducible factor-1α (HIF-1α) can be regulated in transcription, translation, posttranslational modification and other aspects through multiple pathways, and HIF-1α affects multiple metabolic enzymes and membrane proteins during metabolic reprogramming, thus playing a central role in the metabolic reprogramming process, and thus has some implications for tumor therapy and understanding chemotherapy drug resistance. HIF-1α affects a number of metabolic enzymes and membrane proteins in the metabolic reprogramming process, thus playing a central role in the metabolic reprogramming process, which has certain significance for the treatment of tumors and the understanding of chemotherapeutic drug resistance. In this paper, we review the central role of HIF-1α in metabolic reprogramming, chemotherapeutic agents targeting HIF-1α, and chemotherapeutic drug resistance.
Collapse
Affiliation(s)
- Bing Zhu
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| | - Lichao Cheng
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| | - Baosu Huang
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| | - Runzhi Liu
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| | - Bin Ren
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| |
Collapse
|
47
|
Debnath SK, Debnath M, Ghosh A, Srivastava R, Omri A. Targeting Tumor Hypoxia with Nanoparticle-Based Therapies: Challenges, Opportunities, and Clinical Implications. Pharmaceuticals (Basel) 2024; 17:1389. [PMID: 39459028 PMCID: PMC11510357 DOI: 10.3390/ph17101389] [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: 09/21/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Hypoxia is a crucial factor in tumor biology, affecting various solid tumors to different extents. Its influence spans both early and advanced stages of cancer, altering cellular functions and promoting resistance to therapy. Hypoxia reduces the effectiveness of radiotherapy, chemotherapy, and immunotherapy, making it a target for improving therapeutic outcomes. Despite extensive research, gaps persist, necessitating the exploration of new chemical and pharmacological interventions to modulate hypoxia-related pathways. This review discusses the complex pathways involved in hypoxia and the associated pharmacotherapies, highlighting the limitations of current treatments. It emphasizes the potential of nanoparticle-based platforms for delivering anti-hypoxic agents, particularly oxygen (O2), to the tumor microenvironment. Combining anti-hypoxic drugs with conventional cancer therapies shows promise in enhancing remission rates. The intricate relationship between hypoxia and tumor progression necessitates novel therapeutic strategies. Nanoparticle-based delivery systems can significantly improve cancer treatment efficacy by targeting hypoxia-associated pathways. The synergistic effects of combined therapies underscore the importance of multimodal approaches in overcoming hypoxia-mediated resistance. Continued research and innovation in this area hold great potential for advancing cancer therapy and improving patient outcomes.
Collapse
Affiliation(s)
- Sujit Kumar Debnath
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Monalisha Debnath
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Arnab Ghosh
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Rohit Srivastava
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
48
|
Salehpour A, Karimi Z, Ghasemi Zadeh M, Afshar M, Kameli A, Mooseli F, Zare M, Afshar A. Therapeutic potential of mesenchymal stem cell-derived exosomes and miRNAs in neuronal regeneration and rejuvenation in neurological disorders: a mini review. Front Cell Neurosci 2024; 18:1427525. [PMID: 39429946 PMCID: PMC11486650 DOI: 10.3389/fncel.2024.1427525] [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: 05/03/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained considerable attention in the field of regenerative medicine due to their ability to secrete small extracellular vesicles (EVs) known as exosomes. This review delves into the various biological activities of MSCs and the cell interactions enabled by these exosomes, with a focus on their potential for neuronal regeneration and the treatment of neurological disorders. We scrutinize findings from multiple studies that underscore the neuroprotective and neuro-regenerative effects of exosomes derived from MSCs, illuminating their mechanisms of action and therapeutic applications. This review thoroughly investigates all related pathways, miRNAs, and factors to suggest potential strategies for enhancing therapy for neurological disorders using exosomes and miRNAs, and for boosting neuronal regeneration.
Collapse
Affiliation(s)
- Aria Salehpour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zahra Karimi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mokhtar Ghasemi Zadeh
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Afshar
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Kameli
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Fatemeh Mooseli
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Masoud Zare
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Afshar
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
49
|
Huang W, Zhang Z, Li X, Zheng Q, Wu C, Liu L, Chen Y, Zhang J, Jiang X. CD9 promotes TβR2-TβR1 association driving the transition of human dermal fibroblasts to myofibroblast under hypoxia. Mol Med 2024; 30:162. [PMID: 39333849 PMCID: PMC11428569 DOI: 10.1186/s10020-024-00925-5] [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: 03/19/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND During wound healing, fibroblast to myofibroblast transition is required for wound contraction and remodeling. While hypoxia is an important biophysical factor in wound microenvironment, the exact regulatory mechanism underlying hypoxia and fibroblast-to-myofibroblast transition remains unclear. We previously found that tetraspanin CD9 plays an important role in oxygen sensing and wound healing. Herein, we investigated the effects of physiological hypoxia on fibroblast-to-myofibroblast transition and the biological function and mechanism of CD9 in it. METHODS Human skin fibroblasts (HSF) and mouse dermis wounds model were established under physiological hypoxia (2% O2). The cell viability and contractility of HSF under hypoxia were evaluated by CCK8 and collagen gel retraction, respectively. The expression and distribution of fibroblast-to-myofibroblast transition markers and CD9 in HSF were detected by Western blotting and immunofluorescence. CD9 slicing and overexpressing HSFs were constructed to determine the role of CD9 by small interfering RNA and recombinant adenovirus vector. The association of TβR2 and TβR1 was measured by immunoprecipitation to explore the regulatory mechanism. Additionally, further validation was conducted on mouse dermis wounds model through histological analysis. RESULTS Enhanced fibroblast-to-myofibroblast transition and upregulated CD9 expression was observed under hypoxia in vitro and in vivo. Besides, reversal of fibroblast-to-myofibroblast transition under hypoxia was observed when silencing CD9, suggesting that CD9 played a key role in this hypoxia-induced transition. Moreover, hypoxia increased fibroblast-to-myofibroblast transition by activating TGF-β1/Smad2/3 signaling, especially increased interaction of TβR2 and TβR1. Ultimately, CD9 was determined to directly affect TβR1-TβR2 association in hypoxic fibroblast. CONCLUSION Collectively, these findings suggest that CD9 promotes TβR2-TβR1 association, thus driving the transition of human dermal fibroblasts to myofibroblast under hypoxia.
Collapse
Affiliation(s)
- Wanqi Huang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ze Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xin Li
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qingqing Zheng
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Chao Wu
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Luojia Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ying Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Xupin Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
50
|
Deng Y, Shi M, Yi L, Naveed Khan M, Xia Z, Li X. Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment. Heliyon 2024; 10:e37060. [PMID: 39286218 PMCID: PMC11402941 DOI: 10.1016/j.heliyon.2024.e37060] [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: 02/29/2024] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by producing remarkable clinical outcomes for patients with various cancer types. However, only a subset of patients benefits from immunotherapeutic interventions due to the primary and acquired resistance to ICIs. Myeloid-derived suppressor cells (MDSCs) play a crucial role in creating an immunosuppressive tumor microenvironment (TME) and contribute to resistance to immunotherapy. V-domain Ig suppressor of T cell activation (VISTA), a negative immune checkpoint protein highly expressed on MDSCs, presents a promising target for overcoming resistance to current ICIs. This article provides an overview of the evidence supporting VISTA's role in regulating MDSCs in shaping the TME, thus offering insights into how to overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Yayuan Deng
- The First College of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Mengjia Shi
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Yi
- The First College of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Muhammad Naveed Khan
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, 81377, Germany
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Western(Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Park, No. 28 Gaoxin Avenue, High-tech Zone, Chongqing, 401329, China
| |
Collapse
|