1
|
Lei J, Gong D, Duan L, Tang R, Gu W, Zhang F, Zhao Y, Zhang M, Yang X, Yu J. A multidimensional perspective on Poria cocos, an ancient fungal traditional Chinese medicine. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119869. [PMID: 40280371 DOI: 10.1016/j.jep.2025.119869] [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: 08/16/2024] [Revised: 03/25/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Poria cocos occupies a foundational position in Asian medicinal systems, with documented use spanning over 2500 years. Modern applications have expanded its utility across diverse sectors, including functional foods, cosmeceuticals, and veterinary medicine. AIM OF THE WORK This review aims to systematically consolidate historical insights, production, phytochemical profiles, pharmacological mechanisms, and modern applications of Poria cocos, while addressing challenges in quality standardization and sustainable development. MATERIALS AND METHODS For the collection of pertinent information, extensive searches were conducted across diverse literature and electronic databases. Primary keywords such as "Poria cocos" and "Poria" were utilized, supplemented by secondary search terms including "source", "cultivation", "processing", "chemical composition", "pharmacological effects", "practical applications", "quality standard" and "toxicity and safety". Through integration, systematically summarize and analyze the extracted data to evaluate the research progress of Poria cocos. RESULTS This review summarizes the main findings on the history, production, phytochemistry, pharmacology, practical applications, quality standards, and safety of Poria cocos. The medicinal use of Poria cocos has spanned over 2500 years, and its cultivation techniques are constantly evolving. Modern cultivation practices, including pine segment wood and substitute cultivation, highlight trade-offs between yield, cost, and ecological impact. Phytochemical analysis determined that polysaccharides and triterpenes are the main bioactive components, and 120 triterpenoid compounds have been reported. Pharmacological studies have confirmed that Poria cocos has diuretic, liver protective, anti-inflammatory, antitumor, antioxidant, and antidiabetic effects. Practical applications extend beyond medicine to functional foods, cosmeceuticals, and veterinary products. However, there are still challenges in the dynamic balance between resource development and environmental protection, clarification of active ingredients, deepening of pharmacological mechanisms, development of products in practical application fields, and quality standardization, especially in the qualitative and quantitative analysis of active ingredients (polysaccharides and triterpenoids) in Poria cocos, the risk of sulfur fumigation, and heavy metal pollution. Collectively, these findings underscore Poria cocos as a multifaceted resource. CONCLUSION Poria cocos remains a promising candidate for global health and wellness initiatives. Combining traditional knowledge with innovative research to ensure the green cultivation of Poria cocos, with clear ingredients, reliable therapeutic effects, controllable quality, safety and effectiveness, is an important direction for future research.
Collapse
Affiliation(s)
- Juanhong Lei
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Dihong Gong
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Liting Duan
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Ruidan Tang
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Wen Gu
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Fan Zhang
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Yujiao Zhao
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Mei Zhang
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Xingxin Yang
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| | - Jie Yu
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| |
Collapse
|
2
|
Jacinto C, Javed Y, Lavorato G, Tarraga WA, Conde BIC, Orozco JM, Picco AS, Garcia J, Dias CSB, Malik S, Sharma SK. Biotransformation and biological fate of magnetic iron oxide nanoparticles for biomedical research and clinical applications. NANOSCALE ADVANCES 2025; 7:2818-2886. [PMID: 40255989 PMCID: PMC12004083 DOI: 10.1039/d5na00195a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 04/22/2025]
Abstract
Safe implementation of nanotechnology-based products in biomedical applications necessitates an extensive understanding of the (bio)transformations that nanoparticles undergo in living organisms. The long-term fate in the body is a crucial consideration because it governs potential risks for human health. To accurately predict the life cycle of nanoparticles, their fate after administration into the body-including their (bio)transformations, persistence, and biodegradation-needs to be thoroughly evaluated. Magnetic iron oxide nanoparticles (MIONPs) can enter the body through various routes, including inhalation, ingestion, dermal absorption, and injection. Microscale and nanoscale studies are performed to observe nanomaterial biotransformations and their effect on clinically relevant properties. Researchers are utilizing high-resolution TEM for nanoscale monitoring of the nanoparticles while microscale follow-up approaches comprise quantification tools at the whole organism level and the molecular level. Nanoparticle-cell interactions, including cellular uptake and intracellular trafficking, are key to understanding nanoparticle accumulation in cells and organs. Prolonged accumulation may induce cell stress and nanoparticle toxicity, often mediated through oxidative stress and inflammation. In this review article, the journey of nanoparticles in the body is depicted and their biotransformations and final fate are discussed. Immunohistochemical techniques are particularly valuable in tracking nanoparticle distribution within tissues and assessing their impact at the cellular level. A thorough description of a wide range of characterization techniques is provided to unveil the fate and biotransformations of clinically relevant nanoparticles and to assist in their design for successful biomedical applications.
Collapse
Affiliation(s)
- Carlos Jacinto
- Nano-Photonics and Imaging Group, Institute of Physics, Universidade Federal de Alagoas 57072-900 Maceió AL Brazil
| | - Yasir Javed
- Department of Physics, University of Agriculture Faisalabad Pakistan
| | - Gabriel Lavorato
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Wilson A Tarraga
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | | | - Juan Manuel Orozco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Agustin S Picco
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Faculdad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET Diagonal 113 y 64 1900 La Plata Argentina
| | - Joel Garcia
- Department of Chemistry, De La Salle University Manila Philippines
| | - Carlos Sato Baraldi Dias
- Institute for Photon Science and Synchrotron Radiation (IPS), Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 Eggenstein-Leopoldshafen 76344 Germany
| | - Sonia Malik
- Physiology, Ecology & Environmental Laboratory (P2e), University of Orléans 45067 France
- Department of Biotechnology, Baba Farid College Bathinda 151001 India
| | - Surender Kumar Sharma
- Department of Physics, Central University of Punjab Bathinda 151401 India
- Department of Physics, Federal University of Maranhão São Luís 65080-805 Brazil
| |
Collapse
|
3
|
Zhang Q, Chen S, Zhang H, Bao Z, Chen Y, Zhang G, Liu Z, Yang J, He R, Liu Y, Tian X. Optimizing cancer therapy through metal organic frameworks-based nanozymes. Int J Biol Macromol 2025; 306:141409. [PMID: 39993671 DOI: 10.1016/j.ijbiomac.2025.141409] [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: 11/10/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Cancer remains a leading global health challenge, with conventional treatments facing limitations due to drug resistance and adverse effects arising from tumor heterogeneity. Nanozymes, nanomaterials mimicking natural enzymes, have emerged as promising therapeutic agents owing to their catalytic efficiency, stability, and biocompatibility. Among nanozymes, MOFs-based nanozymes are particularly attractive due to the inherent tunability of MOFs, which allows for precise control over their structure, porosity, and catalytic activity. This review comprehensively explores the recent advancements in optimizing cancer therapy through MOFs-based nanozymes. We delve into the classification of these nanozymes based on their enzyme-mimicking activities, including peroxidase, oxidase, catalase, and superoxide dismutase, and discuss their underlying catalytic mechanisms. Additionally, emerging single-atom nanozymes are discussed as a distinct category. Furthermore, we highlight the diverse therapeutic strategies employing MOFs-based nanozymes, such as starvation therapy, oxygen supply, catalytic therapy, glutathione depletion, and activation of therapeutic agents within tumor microenvironment. By exploiting the unique properties of MOFs, these nanozymes demonstrate enhanced therapeutic efficacy in various cancer treatment modalities, including chemotherapy, radiotherapy, photodynamic therapy, and sonodynamic therapy. This review underscores the significant potential of MOFs-based nanozymes as a versatile platform for developing next-generation cancer therapeutics, offering improved targeting, reduced systemic toxicity, and enhanced treatment outcomes.
Collapse
Affiliation(s)
- Qinxin Zhang
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China; Key Laboratory of Carbon Materials of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Sai Chen
- Hebei Key Laboratory of Green Development of Rock and Mineral Materials and Institute of Basalt Fiber Materials, School of Gemmology and Materials Science, Hebei GEO University, Shijiazhuang 050031, China
| | - Hongwei Zhang
- Key Laboratory of Development and Application of Rural Renewable Energy, Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu 610041, China
| | - Zitong Bao
- Key Laboratory of Development and Application of Rural Renewable Energy, Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu 610041, China
| | - Yangyang Chen
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China; Hebei Key Laboratory of Green Development of Rock and Mineral Materials and Institute of Basalt Fiber Materials, School of Gemmology and Materials Science, Hebei GEO University, Shijiazhuang 050031, China
| | - Guangling Zhang
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China
| | - Zhiyong Liu
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China
| | - Jichun Yang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Runhe He
- Key Laboratory of Carbon Materials of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Yatao Liu
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xuetao Tian
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China.
| |
Collapse
|
4
|
Ham O, Jung S, Kim YJ, Woo DH, Jeong JS, Kim W, Kim S, Quah Y, Lee S, Yu WJ. Methotrexate-induced disruption of the serum-glucocorticoid inducible protein kinase 3 signaling pathway and its effects on brain development. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138438. [PMID: 40327939 DOI: 10.1016/j.jhazmat.2025.138438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Methotrexate (MTX) is a widely used chemotherapeutic and immunosuppressive agent. It is frequently detected in the environment due to its low biodegradability and toxic properties, which may pose risk to humans via contaminated drinking water. MTX impacts brain development; however, the underlying mechanisms remain poorly understood. In this study, we aimed to investigate the potential of brain cortical organoids (COs) to exhibit developmental neurotoxicity upon exposure to MTX and elucidated the underlying mechanisms. We showed that MTX affected brain development by increasing reactive oxygen species production, endoplasmic reticulum stress, and apoptosis in COs, affecting immature neurons, gamma-aminobutyric acid (GABA)ergic neurons, and astrocytes. Treatment with folic acid ameliorated the MTX-induced cellular damage. Furthermore, RNA-seq analysis revealed the significant downregulation of serum/glucocorticoid-regulated kinase family member 3 (SGK3) by MTX treatment. Further investigation using RNA interference with an siRNA targeting SGK3 confirmed that the SGK3 signaling pathway plays an essential role in regulating brain development. Finally, we demonstrated calcium homeostasis disruption in MTX-treated COs (MTX-COs). Our findings suggest that MTX affects cortical development in the brain by increasing intracellular ROS and ER stress, ultimately inducing apoptosis through the inhibition of the SGK3 signaling pathway in MTX-COs. Furthermore, abnormal brain development can be caused by an imbalance in intracellular calcium homeostasis and alterations in the neuronal cell expression in MTX-COs. These results indicate that MTX causes developmental neurotoxicity and underscore the importance of informed decision making regarding the clinical use of MTX, particularly during pregnancy.
Collapse
Affiliation(s)
- Onju Ham
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea.
| | - Soontag Jung
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - Ye-Ji Kim
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Dong Ho Woo
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Ji-Seong Jeong
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - Woojin Kim
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - Sangyun Kim
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - Yixian Quah
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - SeungJin Lee
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea
| | - Wook-Joon Yu
- Center for Regulatory Toxicology Research, Laboratory of Developmental and Reproductive Toxicology, Korea Institute of Toxicology, Deajeon 34114, Republic of Korea.
| |
Collapse
|
5
|
Pervez A, Khan B, Khan GN, Khattak S, Ali M, Mujeeb K, Nasib B, Kim HG, Qureshi IZ, Arshad M. Evaluation of hepatic cancer stem cells (CD 73+, CD 44+, and CD 90+) induced by diethylnitrosamine in male rats and treatment with biologically synthesized silver nanoparticles. Mol Biol Rep 2025; 52:393. [PMID: 40232523 DOI: 10.1007/s11033-025-10495-2] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Cancer stem cells (CSCs) play a critical role in the initiation and heterogeneity of a variety of cancers due to their pluripotent nature and capacity for asymmetric cell division. Therefore, uncovering the carcinogens that increase the CSC population in target tissues is crucial for understanding the genesis of cancer. The therapeutic potential of Operculina turpethum (OT) derived silver nanoparticles (AgNPs) was assessed in diethylnitrosamine (DEN)-induced CSC populations; CD73+, CD44+, and CD90 + of hepatic tissues in male rats. METHODS Histopathology, fluorescence-activated cell sorting (FACS), and RT-qPCR were performed on the control, DEN, DEN + AgNPs, and AgNPs-treated groups. AgNPs were characterized by FTIR, EDX, XRD, and SEM. RESULTS AgNPs were confirmed by intense surface plasmon resonance at 425 nm. Antioxidants, the reducing sugars responsible for Ag+ 1 reduction and subsequent conjugate formation with nanoparticles, were confirmed by vibrational spectra. The spherical morphology, composition, and conjugation of silver nanoparticles to phytoconstituents with partially crystalline, face-centered cubic structure were established through SEM, EDX spectrum, and XRD, respectively. Disrupted tissue architecture, cell enlargement, mild pleomorphism, and expanded central veins were observed in hepatic tissues of DEN-treated animals. However, a moderate inflammatory response was observed in the DEN + AgNPs-treated group. CSC populations were significantly increased in the DEN-treated group, but decreased with AgNPs-treatment. The mRNA expression levels of CD90, CD44, and CD73 genes were significantly up-regulated in the DEN-treated group compared to control group however, in DEN + AgNPs and AgNPs groups it were similar to control group. CONCLUSION All together, DEN-induced the hepatic CSC cell populations and the OT mediated AgNPs have therapeutic potential to attenuate the harmful effects of DEN. This study provides evidenced that OT-synthesised AgNPs may be considered as a therapeutic agent for liver related malignancies.
Collapse
Affiliation(s)
- Amber Pervez
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Behramand Khan
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Gul Nabi Khan
- Department of Zoology, Islamia College University, Peshawar, Pakistan.
| | - Sumayya Khattak
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Mazhar Ali
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Komal Mujeeb
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Bushra Nasib
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Hyung Goo Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, The State University of New Jersey, Rutgers, USA
| | | | - Muhammad Arshad
- Biochemistry Section, Jhang Campus, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| |
Collapse
|
6
|
Yuan L, Cai Y, Wang G, Liu X, Chen B, Zhou D, Wu Y, Qu N, Li X, Zhou W. SGK3 promotes estrogen receptor-positive breast cancer proliferation by activating STAT3/ZMIZ2 pathway to stabilise β-catenin. Br J Pharmacol 2025; 182:1856-1875. [PMID: 39876548 DOI: 10.1111/bph.17453] [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: 05/17/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer is a leading threat to women's health, with approximately 70% of cases being estrogen receptor-positive. SGK3 is regulated by estrogen and is positively associated with estrogen receptor expression, although its molecular role remains unclear. EXPERIMENTAL APPROACH Proteomics was used to identify SGK3's downstream targets. Tissue microarray immunofluorescence evaluated SGK3 and ZMIZ2 expression in ER+ breast cancer. Lentiviral-mediated knockdown and overexpression of SGK3 and/or ZMIZ2 assessed their effects on cell proliferation in vitro and in vivo. Chromatin immunoprecipitation (ChIP) analyzed p-STAT3 binding to the ZMIZ2 promoter, and Co-immunoprecipitation (Co-IP) examined ZMIZ2-β-catenin interaction. KEY RESULTS SGK3 expression was elevated in breast tumour tissues correlating with reduced patient survival. Proteomic analysis identified ZMIZ2 as a downstream target of SGK3. Overexpression of SGK3 promoted the proliferation of estrogen receptor-positive breast cancer in MCF-7 and T47D cells. Inhibition had the opposite effects. ZMIZ2 overexpression rescued the proliferation deficit in SGK3 knockdown cells. ZMIZ2 was found to bind and stabilises β-catenin. Knockdown of SGK3 led to β-catenin degradation via polyubiquitination, a process reversed by ZMIZ2 overexpression. STAT3 was identified as a downstream effector of SGK3 and its knockdown reduced cytoplasmic and nuclear p-STAT3 and STAT3, and inhibited ZMIZ2 and β-catenin expression. Celastrol suppressed estrogen receptor-positive breast cancer cell proliferation by inhibiting the SGK3/STAT3/ZMIZ2/β-catenin pathway. CONCLUSIONS AND IMPLICATIONS SGK3 expression is associated with poorer survival rates, thus SGK3 is a potential therapeutic target. As celastrol can inhibit SGK3 expression it could be an effective therapeutic agent.
Collapse
Affiliation(s)
- Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Pharmacy, The Third People's Hospital of Chengdu, Sichuan, China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Duanfang Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Wu G, Zhong C, Tian X, Zha L, Hou L, Feng X. Emerging roles of hyaluronic acid hydrogels in cancer treatment and wound healing: A review. Int J Biol Macromol 2025; 303:140442. [PMID: 39880244 DOI: 10.1016/j.ijbiomac.2025.140442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
Hyaluronic acid (HA)-derived hydrogels demonstrate a significant development in the biomedical uses, especially in cancer treatment and wound repair. Cancer continues to be one of the leading causes of death worldwide, with current therapies frequently impeded by lack of specificity, side effects, and the emergence of resistance. HA hydrogels, characterized by their distinctive three-dimensional structure, hydrophilic nature, and biocompatibility, develop an advanced platform for precise drug delivery, improving therapeutic results while minimizing systemic toxicity. These hydrogels facilitate the controlled release of drugs, genes, and various therapeutic substances, enhancing the effectiveness of chemotherapy, radiotherapy, and immunotherapy. Additionally, they can be designed to react to stimuli such as pH, light, and magnetic fields, enhancing their therapeutic capabilities. In the process of wound healing, the hydrophilic and porous characteristics of HA hydrogels establish a moist environment encouraging cell growth and contributes to the tissue recovery. By imitating the extracellular matrix, they promote tissue regeneration, improve angiogenesis, and influence immune reactions. This review examines the various functions of HA-based hydrogels in cancer treatment and wound healing, highlighting their advancement, applications, and ability to change existing therapeutic methods in these important health sectors.
Collapse
Affiliation(s)
- Gang Wu
- Department of Hepatobiliary Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chunyan Zhong
- Department of Ultrasound, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xiaohui Tian
- Department of Obstetrics and Gynecology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Lisha Zha
- School of Biomedical Sciences, Hunan University, Changsha, 410082, Hunan, PR China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu 610041, Sichuan, China.
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, No. 89 Xiguan Road, Gaozhou 525299, Guangdong, China.
| |
Collapse
|
8
|
Akbari Oryani M, Tarin M, Rahnama Araghi L, Rastin F, Javid H, Hashemzadeh A, Karimi-Shahri M. Synergistic cancer treatment using porphyrin-based metal-organic Frameworks for photodynamic and photothermal therapy. J Drug Target 2025; 33:473-491. [PMID: 39618308 DOI: 10.1080/1061186x.2024.2433551] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 02/25/2025]
Abstract
Recent advancements in multifunctional nanomaterials for cancer therapy have highlighted porphyrin-based metal-organic frameworks (MOFs) as promising candidates due to their unique properties and versatile applications. This overview focuses on the use of porphyrin-based MOFs for combined photodynamic therapy (PDT) and photothermal therapy (PTT) in cancer treatment. Porphyrin-based MOFs offer high porosity, tuneable structures, and excellent stability, making them ideal for drug delivery and therapeutic applications. The incorporation of porphyrin molecules into the MOF framework enhances light absorption and energy transfer, leading to improved photodynamic and photothermal effects. Additionally, the porosity of MOFs allows for the encapsulation of therapeutic agents, further enhancing efficacy. In PDT, porphyrin-based MOFs generate reactive oxygen species (ROS) upon light activation, destroying cancer cells. The photothermal properties enable the conversion of light energy into heat, resulting in localised hyperthermia and tumour ablation. The combination of PDT and PTT in a single platform offers synergistic effects, leading to better therapeutic outcomes, reduced side effects, and improved selectivity. This dual-modal treatment strategy provides precise spatiotemporal control over the treatment process, paving the way for next-generation therapeutics with enhanced efficacy and reduced side effects. Further research and optimisation are needed for clinical applications.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Leila Rahnama Araghi
- Department of Biotechnology, Faculty of Science, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farangis Rastin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
9
|
Zhang G, Jiang X, Xia Y, Qi P, Li J, Wang L, Wang Z, Tian X. Hyaluronic acid-conjugated lipid nanocarriers in advancing cancer therapy: A review. Int J Biol Macromol 2025; 299:140146. [PMID: 39842601 DOI: 10.1016/j.ijbiomac.2025.140146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/03/2025] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Lipid nanoparticles are obtaining significant attention in cancer treatment because of their efficacy at delivering drugs and reducing side effects. These things are like a flexible platform for getting anticancer drugs to the tumor site, especially upon HA modification, a polymer that is known to target tumors overexpressing CD44. HA is promising in cancer therapy because it taregtes tumor cells by binding onto CD44 receptors, which are often upregulated in cancer cells. Lipid nanoparticles are not only beneficial in improving solubility and stability of drugs; they also use the EPR effect, meaning they accumulate more in tumor tissue than in healthy tissue. Adding HA to these nanoparticles expands their biocompatibility and makes them more accurate and specific towards tumor cells. Studies show that HA-modified nanoparticles carrying drugs such as paclitaxel or doxorubicin improve how well cells absorb the drugs, reduce drug resistance, and make tumor shrinking. These nanoparticles can respond to tumor microenvironment stimuli in targeted delivery. This targeted delivery diminishes side effects and improves anti-cancer activity of drugs. Thus, lipid-based nanoparticles conjugated with HA are a promising way to treat cancer by delivering drugs effectively, minimizing side effects, and giving us better therapeutic results.
Collapse
Affiliation(s)
- Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, China
| | - Xin Jiang
- Department of Clinical Pharmacy, Baoying People's Hospital, Affiliated Hospital of Medical School, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yitong Xia
- Department of Oral Medicine, Jining Medical College, Jining, Shandong, China
| | - Pengpeng Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jie Li
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, China
| | - Lizhen Wang
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan City, Shandong, China.
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng City Hospital of Traditional Chinese Medicine, Liaocheng, Shandong, China.
| | - Xiuli Tian
- Department of Respiration, Liaocheng People's Hospital, Liaocheng, Shandong, China.
| |
Collapse
|
10
|
Wang R, Wang Q, Liao J, Yu X, Li W. Piperlongumine overcomes osimertinib resistance via governing ubiquitination-modulated Sp1 turnover. JCI Insight 2025; 10:e186165. [PMID: 40125551 PMCID: PMC11949057 DOI: 10.1172/jci.insight.186165] [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: 08/19/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common cause of cancer-related deaths worldwide, and its incidence has been increasing in recent years. While targeted therapies like osimertinib, an epidermal growth factor receptor tyrosine kinase inhibitor, have brought about notable improvements in patient outcomes for advanced NSCLC, the challenge of acquired drug resistance persists. Here, we found that cellular mesenchymal-epithelial transition factor (c-Met) was highly expressed in osimertinib-resistant cells, and depletion of c-Met markedly inhibited the growth of osimertinib-resistant cells ex vivo and in vivo, suggesting that c-Met is a potential target to address osimertinib resistance. Through a screening process using a natural product compound library, we identified piperlongumine as a potent inhibitor to overcome osimertinib resistance. Furthermore, the combined treatment of piperlongumine and osimertinib exhibited robust antitumor effects in resistant cells, partially restoring their sensitivity to osimertinib. Additionally, we discovered that piperlongumine could enhance the interaction between E3 ligase RNF4 and Sp1, inhibit the phosphorylation of Sp1 at Thr739, facilitate the ubiquitination and degradation of Sp1, lead to c-Met destabilization, and trigger intrinsic apoptosis in resistant cells. In summary, our study sheds light on the potential of piperlongumine in overcoming osimertinib resistance, offering new strategies and perspectives for the clinical management of drug-resistant NSCLC.
Collapse
Affiliation(s)
| | - Qiang Wang
- NHC Key Laboratory of Translational Research on Transplantation Medicine, Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | | | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Li
- Department of Radiology and
| |
Collapse
|
11
|
Kang T, Wang Y, Jiang Y, Chen S, Lin N, Guo M, Zhu H, Tang D, Ding X, He M. The SGK3/GSK3β/β-catenin signaling promotes breast cancer stemness and confers resistance to alpelisib therapy. Int J Biol Sci 2025; 21:2462-2475. [PMID: 40303291 PMCID: PMC12035905 DOI: 10.7150/ijbs.104850] [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: 10/08/2024] [Accepted: 03/09/2025] [Indexed: 05/02/2025] Open
Abstract
Drug resistance is the leading cause of death in patients with advanced tumors. Alpelisib, a selective PIK3CA inhibitor, has been recently approved for treating advanced breast cancer. However, drug resistance is inevitable, and the mechanisms behind alpelisib-associated resistance remain elusive. To address this problem, we established an alpelisib-resistant breast cancer cell model and confirmed that the resistant cells exhibited enhanced abilities in colony formation, migration, anti-apoptosis, spheroidization, tumor formation and metastasis. Further analysis revealed that SGK3 was significantly upregulated in alpelisib-resistant cells, which was strongly associated with tumor stemness. Additionally, we observed that SGK3 promoted tumor cell stemness by activating GSK3β/β-catenin signaling pathway, leading to the resistance to alpelisib in breast cancer. Finally, we demonstrated that SGK3 inhibitor could restore the sensitivity of resistant cells to alpelisib. Collectively, these findings suggest that SGK3 could be a novel therapeutic target for breast cancer patients who developed resistance to alpelisib.
Collapse
Affiliation(s)
- Tingting Kang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yuanfang Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yaxin Jiang
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shunjie Chen
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Na Lin
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Minyan Guo
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Haotu Zhu
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Di Tang
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaofan Ding
- Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Mian He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
12
|
Kobia FM, Castro E Almeida L, Paganoni AJ, Carminati F, Andronache A, Lavezzari F, Wade M, Vaccari T. Novel determinants of NOTCH1 trafficking and signaling in breast epithelial cells. Life Sci Alliance 2025; 8:e202403122. [PMID: 39663000 PMCID: PMC11633778 DOI: 10.26508/lsa.202403122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
The evolutionarily conserved Notch signaling pathway controls cell-cell communication, enacting cell fate decisions during development and tissue homeostasis. Its dysregulation is associated with a wide range of diseases, including congenital disorders and cancers. Signaling outputs depend on maturation of Notch receptors and trafficking to the plasma membrane, endocytic uptake and sorting, lysosomal and proteasomal degradation, and ligand-dependent and independent proteolytic cleavages. We devised assays to follow quantitatively the trafficking and signaling of endogenous human NOTCH1 receptor in breast epithelial cells in culture. Based on such analyses, we executed a high-content screen of 2,749 human genes to identify new regulators of Notch that might be amenable to pharmacologic intervention. We uncovered 39 new NOTCH1 modulators for NOTCH1 trafficking and signaling. Among them, we find that PTPN23 and HCN2 act as positive NOTCH1 regulators by promoting endocytic trafficking and NOTCH1 maturation in the Golgi apparatus, respectively, whereas SGK3 serves as a negative regulator that can be modulated by pharmacologic inhibition. Our findings might be relevant in the search of new strategies to counteract pathologic Notch signaling.
Collapse
Affiliation(s)
- Francis M Kobia
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | | | - Alyssa Jj Paganoni
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | | | - Adrian Andronache
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | | | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Thomas Vaccari
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
13
|
Feng JH, Zhang ML, Zou YM, Tang XY, Chen XT, Meng W, Chen M, Li RT, Chen JX. Hollow gold-platinum nanoshells as a delivery platform for Ce6: cascading catalysis for enhanced multimodal therapy in tumor ablation and antitumor immunity. NANOSCALE 2025; 17:5456-5471. [PMID: 39901668 DOI: 10.1039/d4nr04627g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Precious metal nanozymes are renowned for their enzyme-mimicking properties, which can modulate the tumor microenvironment (TME) and enhance treatment. However, their small size often leads to aggregation and their single and limited catalytic potential impedes antitumor and immune-activating capabilities. To address these limitations, we developed a nanocomposite with multiple enzyme activities that synergistically enhances photodynamic and photothermal therapy (PDT and PTT), significantly boosting antitumor efficacy and immune response. Our approach involved using UiO-66-NH2 to facilitate the growth of gold-platinum bimetallic nanozymes, resulting in a core-shell structure of UiO-66-NH2@AuPt (UAuPt). The UiO-66-NH2 was then etched to create hollow gold-platinum bimetallic (HAuPt) nanoshells and further encapsulated with PEG-SH and the photosensitizer Ce6 to form the HAuPt@Ce6-PEG-SH (HCP) nanocomposite. Regarding the HCP nanocomposite, its absorption capability in the near-infrared second (NIR-II) region makes it a suitable photothermal agent for PTT, while Ce6 serves as the active agent for PDT. Furthermore, the gold nanoparticles (Au NPs) and platinum nanoparticles (Pt NPs) exhibit glucose oxidase (GOD)-, catalase (CAT)-, and peroxidase (POD)-like activities. This triple-enzyme activity forms an efficient cascade catalytic system, leading to refined remodeling of the TME and efficient enhancement of PTT and PDT. Moreover, the combination therapy triggers tumor-associated macrophage (TAM) polarization and immunogenic cell death (ICD), which not only promotes dendritic cell (DC) maturation but also stimulates T cell activation and the release of tumor-specific immune factors. This cascade ultimately results in a robust antitumor immune response. The in vitro and in vivo results demonstrated a significant antitumor efficacy and immune response, promising efficient nanozymes for therapeutic advancement.
Collapse
Affiliation(s)
- Jia-Hao Feng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Mei-Lian Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yi-Ming Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Xiao-Yan Tang
- School of Chemistry and Materials Engineering, Jiangsu Key Laboratory of Advanced Functional Materials, Changshu Institute of Technology, Changshu 215500, China
| | - Xiao-Tong Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Wei Meng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Ming Chen
- The People's Hospital of Gaozhou, Gaozhou, Guangdong 525200, China.
| | - Rong-Tian Li
- Department of Clinical Pharmacy, Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China.
| | - Jin-Xiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
14
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
15
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [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: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
16
|
Zhou X, Feng S, Xu Q, Li Y, Lan J, Wang Z, Ding Y, Wang S, Zhao Q. Current advances in nanozyme-based nanodynamic therapies for cancer. Acta Biomater 2025; 191:1-28. [PMID: 39571955 DOI: 10.1016/j.actbio.2024.11.023] [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: 08/16/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Nanozymes are nano-catalysis materials with enzyme-like activities, which can repair the defects of natural enzyme such as harsh catalytic conditions, and harness their strengths to treat tumor. The emerging nanodynamic therapies improved drug selectivity and decreased drug tolerance, while causing efficient cell apoptosis through the generated reactive oxygen species (ROS). Nanodynamic therapies based on nanozymes can improve the complicated tumor microenvironment (TME) to reduce the defect rate of nanodynamic therapies, and provide more options for tumor treatment. This review summarized the characteristics and applications of nanozymes with different activities and the factors influencing the activity of nanozymes. We also focused on the application of nanozymes in nanodynamic therapies, including photodynamic therapy (PDT), chemodynamic therapy (CDT), and sonodynamic therapy (SDT). Moreover, we discussed the strategies for optimizing nanodynamic therapies based on nanozymes for tumor treatment in detail, and provided a systematic review of tactics for synergies with other tumor therapies. Ultimately, we analyzed the shortcomings of nanodynamic therapies based on nanozymes and the relevant research prospect, which would provide sufficient evidence and lay a foundation for further research. STATEMENT OF SIGNIFICANCE: 1. The novelty and significance of the work with respect to the existing literatures. (1) Recent advances in nanozyme-based nanodynamic therapies are comprehensively and systematically reviewed, and strategies to address the limitations and challenges of current therapies based on nanozymes are discussed firstly. (2) The mechanism of nanozymes in nanodynamic therapies is described for the first time. The synergistic therapies, prospects, and challenges of nanozyme-based nanodynamic therapies are innovatively discussed. 2. The scientific impact and interest to our readership. This review focuses on the recent progress of nanozyme-based nanodynamic therapies. This review indicates the way forward for the combined treatment of nanozymes and nanodynamic therapies, and lays a foundation for facilitating theoretical development in clinic.
Collapse
Affiliation(s)
- Xubin Zhou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Shuaipeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yian Li
- School of Libra Arts of Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jiaru Lan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Ziyi Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yiduo Ding
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
17
|
Luo J, Huang H, Jiang J, Zheng W, Chen P, Bai H. Clearance of Intracellular Pathogens with Hyaluronic Acid Nanomicelles Responsive to H 2S and pH. Molecules 2024; 29:5971. [PMID: 39770059 PMCID: PMC11678907 DOI: 10.3390/molecules29245971] [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: 11/07/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Hyaluronic acid (HA) is an acidic mucopolysaccharide of animal origin composed of repeating disaccharide units of N-acetylglucosamine and glucuronic acid. Due to its excellent biocompatibility, biodegradability, and selective affinity for CD44 receptors on cell surfaces, HA is widely employed as a drug carrier. In our study, we aimed to target subcellular bacteria by grafting cystamine onto HA scaffolds through an amide reaction, producing a linker responsive to H2S and pH changes. Subsequently, hydrophobic dodecylamine was attached to HA, forming amphiphilic molecules. These amphiphilic entities can self-assemble into nanomicelles in an aqueous solution, thereby encapsulating the antibacterial agent triclosan (TCS). The resulting HA-based system (HASS-TCS) can be internalized via CD44-mediated endocytosis, releasing substantial amounts of streptomycin and TCS in H2S-rich and acidic environments. Additionally, HASS-TCS has demonstrated effectiveness in eradicating biofilms and addressing intracellular infections caused by Salmonella. This study underscores a novel pH-sensitive hyaluronic acid-based drug delivery system with significant potential for the effective treatment of intracellular infections.
Collapse
Affiliation(s)
| | | | | | | | - Peng Chen
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang, Tarim University, Alar 843300, China; (J.L.); (H.H.); (J.J.); (W.Z.)
| | - Hongjin Bai
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang, Tarim University, Alar 843300, China; (J.L.); (H.H.); (J.J.); (W.Z.)
| |
Collapse
|
18
|
Rumon MM, Akib AA, Sarkar SD, Khan MAR, Uddin MM, Nasrin D, Roy CK. Polysaccharide-Based Hydrogels for Advanced Biomedical Engineering Applications. ACS POLYMERS AU 2024; 4:463-486. [PMID: 39679058 PMCID: PMC11638789 DOI: 10.1021/acspolymersau.4c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 12/17/2024]
Abstract
In recent years, numerous applications of hydrogels using polysaccharides have evolved, benefiting from their widespread availability, excellent biodegradability, biocompatibility, and nonpoisonous nature. These natural polymers are typically sourced from renewable materials or from manufacturing processes, contributing collaboratively to waste management and demonstrating the potential for enhanced and enduring sustainability. In the field of novel bioactive molecule carriers for biotherapeutics, natural polymers are attracting attention due to their inherent properties and adaptable chemical structures. These polymers offer versatile matrices with a range of architectures and mechanical properties, while retaining the bioactivity of incorporated biomolecules. However, conventional polysaccharide-based hydrogels suffer from inadequate mechanical toughness with large swelling properties, which prohibit their efficacy in real-world applications. This review offers insights into the latest advancements in the development of diverse polysaccharide-based hydrogels for biotherapeutic administrations, either standalone or in conjunction with other polymers or drug delivery systems, in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- Md. Mahamudul
Hasan Rumon
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
| | - Anwarul Azim Akib
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
| | - Stephen Don Sarkar
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United
States
| | | | - Md. Mosfeq Uddin
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
- Department
of Chemistry, University of Victoria, Victoria 3800, Canada
| | - Dina Nasrin
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
| | - Chanchal Kumar Roy
- Department
of Chemistry, Bangladesh University of Engineering
and Technology, Dhaka 1000, Bangladesh
| |
Collapse
|
19
|
Zeng F, Chen Y, Lin J. Identification of alternative lengthening of telomeres-related genes prognosis model in hepatocellular carcinoma. BMC Cancer 2024; 24:1386. [PMID: 39529015 PMCID: PMC11555837 DOI: 10.1186/s12885-024-13146-0] [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: 07/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor worldwide, characterized by high mortality. This study aimed to explore the prognostic value and function of alternative lengthening of telomeres (ALT)-related genes in HCC. METHODS Differentially expressed genes (DEGs) were identified based on The Cancer Genome Atlas (TCGA) and then intersected with ALT-related genes to obtain ALTDEGs. Risk score model was constructed using the least absolute shrinkage and selection operator (LASSO) algorithm and Cox regression and validated with Gene Expression Omnibus (GEO) datasets. The predictive efficacy of the risk score and ALTs-score was evaluated by Kaplan-Meier curves, time-ROC curves, and the nomogram analyses. The impacts of SMG5 silencing on the HCC cell behaviors were assessed by CCK-8, wound healing, and Transwell assays. RESULTS A total of 500 ALTDEGs were screened and 13 genes (CDCA8, SMG5, RAD54B, FOXD2, NOL10, RRP12, CCT5, CCT4, HDAC1, DDX1, HRG, HDAC2, and PPP1CB) were identified for constructing a prognostic model. The overall survival (OS) curves, time-ROC curves, and nomograms based on the risk score or ALTs-score were developed to optimally predict the survival of HCC patients. ALTs-score was correlated with immune infiltration and confirmed its value in predicting immunotherapy outcomes. Furthermore, RT-qPCR demonstrated that eight risk signature genes were up-regulated in HCC cells. SMG5 silencing suppressed the proliferation, migration, and invasion of HCC cells. It was also found that SMG5 silencing reduced C-circle level in SNU-387 cells. CONCLUSION We identified new ALT-related prognostic biomarkers for HCC. SMG5 knockdown inhibited the HCC progression, which might be a promising target for HCC therapy.
Collapse
Affiliation(s)
- FanLin Zeng
- Department of General Surgery (Hepatobiliary and Pancreatic Surgery), The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - YuLiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jie Lin
- Department of Intensive Medicine (Comprehensive Intensive Care Unit), The First Affiliated Hospital of Gannan Medical University, No. 128 Jin Ling Lu, Ganzhou, Jiangxi, 341000, P.R. China.
| |
Collapse
|
20
|
Wang D, Yao H, Ye J, Gao Y, Cong H, Yu B. Metal-Organic Frameworks (MOFs): Classification, Synthesis, Modification, and Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404350. [PMID: 39149999 DOI: 10.1002/smll.202404350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Metal-organic frameworks (MOFs) are a new variety of solid crystalline porous functional materials. As an extension of inorganic porous materials, it has made important progress in preparation and application. MOFs are widely used in various fields such as gas adsorption storage, drug delivery, sensing, and biological imaging due to their high specific surface area, porosity, adjustable pore size, abundant active sites, and functional modification by introducing groups. In this paper, the types of MOFs are classified, and the synthesis methods and functional modification mechanisms of MOFs materials are summarized. Finally, the application prospects and challenges of metal-organic framework materials in the biomedical field are discussed, hoping to promote their application in multidisciplinary fields.
Collapse
Affiliation(s)
- Dayang Wang
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Huanchen Yao
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Jiashuo Ye
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yan Gao
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
- School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
21
|
Chen Y, Wang C, Hu S, Liu X. HRS Facilitates Newcastle Disease Virus Replication in Tumor Cells by Promoting Viral Budding. Int J Mol Sci 2024; 25:10060. [PMID: 39337546 PMCID: PMC11432301 DOI: 10.3390/ijms251810060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Newcastle disease virus (NDV) is a highly pathogenic avian infectious disease agent and also a promising oncolytic virus with broad application prospects. The Endosomal Sorting Complex Required for Transport (ESCRT) machinery has been increasingly recognized for its crucial role in the life cycles of enveloped viruses, influencing processes such as viral entry, replication, and budding. In this study, we employed an RNA interference screening approach to identify key ESCRT components that regulate NDV replication in tumor cells. qPCR, immunofluorescence, and Western blot assays demonstrated that knockdown of HRS, CHMP4A, CHMP4B, and CHMP4C significantly impaired NDV replication in HeLa cells, with HRS exhibiting the most pronounced inhibitory effect. Additionally, HRS knockout significantly inhibited viral budding and suppressed NDV-induced cell death in HeLa cells. Notably, NDV infection was shown to significantly upregulate HRS gene and protein expression in a time-dependent manner. In conclusion, this study systematically identifies critical ESCRT components involved in NDV replication within tumor cells, with a particular focus on the role of HRS in promoting NDV's replication by promoting viral budding, offering new insights for the development of NDV-based oncolytic therapies.
Collapse
Affiliation(s)
- Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| | - Chunxuan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| |
Collapse
|
22
|
Dowaidar M. Cell-penetrating peptides with nanoparticles hybrid delivery vectors and their uptake pathways. Mitochondrion 2024; 78:101906. [PMID: 38797356 DOI: 10.1016/j.mito.2024.101906] [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: 03/04/2024] [Revised: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Cell-penetrating peptides (CPPs) are molecules that improve the cellular uptake of various molecular payloads that do not easily traverse the cellular membrane. CPPs can be found in pharmaceutical and medical products. The vast majority of cell-penetrating chemicals that are discussed in published research are peptide based. The paper also delves into the various applications of hybrid vectors. Because CPPs are able to carry cargo across the cellular membrane, they are a viable candidate for use as a suitable carrier for a wide variety of cargoes, such as siRNA, nanoparticles, and others. In which we discuss the CPPs, their classification, uptake mechanisms, hybrid vector systems, nanoparticles and their uptake mechanisms, etc. Further in this paper, we discuss CPPs conjugated to Nanoparticles, Combining CPPs with lipids and polymeric Nanoparticles in A Conjugated System, CPPs conjugated to nanoparticles for therapeutic purposes, and potential therapeutic uses of CPPs as delivery molecules. Also discussed the preclinical and clinical use of CPPS, intracellular trafficking of nanoparticles, and activatable and bioconjugated CPPs.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia.
| |
Collapse
|
23
|
Ren J, Che Y, Li H, Gao H, Wang Y, Wang Y, Su H, Li Z, Li J, Qu P. SGK3 deficiency in macrophages suppresses angiotensin II-induced cardiac remodeling via regulating Ndufa13-mediated mitochondrial oxidative stress. Cell Mol Life Sci 2024; 81:359. [PMID: 39158709 PMCID: PMC11335188 DOI: 10.1007/s00018-024-05395-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: 04/04/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
Infiltration of monocyte-derived macrophages plays a crucial role in cardiac remodeling and dysfunction. The serum and glucocorticoid-inducible protein kinase 3 (SGK3) is a downstream factor of PI3K signaling, regulating various biological processes via an AKT-independent signaling pathway. SGK3 has been implicated in cardiac remodeling. However, the contribution of macrophagic SGK3 to hypertensive cardiac remodeling remains unclear. A cardiac remodeling model was established by angiotensin II (Ang II) infusion in SGK3-Lyz2-CRE (f/f, +) and wild-type mice to assess the function of macrophagic SGK3. Additionally, a co-culture system of SGK3-deficient or wild-type macrophages and neonatal rat cardiomyocytes (CMs) or neonatal rat fibroblasts (CFs) was established to evaluate the effects of SGK3 and the underlying mechanisms. SGK3 levels were significantly elevated in both peripheral blood mononuclear cells and serum from patients with heart failure. Macrophage SGK3 deficiency attenuated Ang II-induced macrophage infiltration, myocardial hypertrophy, myocardial fibrosis, and mitochondrial oxidative stress. RNA sequencing suggested Ndufa13 as the candidate gene in the effect of SGK3 on Ang II-induced cardiac remolding. Downregulation of Ndufa13 in CMs and CFs prevented the suppression of cardiac remodeling caused by SGK3 deficiency in macrophages. Mechanistically, the absence of SGK3 led to a reduction in IL-1β secretion by inhibiting the NLRP3/Caspase-1/IL-1β pathway in macrophages, consequently suppressing upregulated Ndufa13 expression and mitochondrial oxidative stress in CMs and CFs. This study provides new evidence that SGK3 is a potent contributor to the pathogenesis of hypertensive cardiac remodeling, and targeting SGK3 in macrophages may serve as a potential therapy for cardiac remodeling.
Collapse
Affiliation(s)
- Jiayu Ren
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Yilin Che
- The 1st Department of Thoracic Medical Oncology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Heyu Li
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Haijun Gao
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Yue Wang
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Ying Wang
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Hongtong Su
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China
| | - Zhihan Li
- The Department of Pathology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Li
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China.
| | - Peng Qu
- Department of Cardiology, Institute of Heart and Vascular Diseases, Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, P.R. China.
- Faculty of Medicine, Dalian University of Technology, No.2 Linggong Road, Ganjingzi District, Liaoning, 116024, P.R. China.
| |
Collapse
|
24
|
Agwa MM, Marzouk RE, Sabra SA. Advances in active targeting of ligand-directed polymeric nanomicelles via exploiting overexpressed cellular receptors for precise nanomedicine. RSC Adv 2024; 14:23520-23542. [PMID: 39071479 PMCID: PMC11273262 DOI: 10.1039/d4ra04069d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Many of the utilized drugs which already exist in the pharmaceutical sector are hydrophobic in nature. These drugs are characterized by being poorly absorbed and difficult to formulate in aqueous environments with low bioavailability, which could result in consuming high and frequent doses in order to fulfil the required therapeutic effect. As a result, there is a decisive demand to find modern alternatives to overcome all these drawbacks. Self-assembling polymeric nanomicelles (PMs) with their unique structure appear to be a fascinating choice as a pharmaceutical carrier system for improving the solubility & bioavailability of many drugs. PMs as drug carriers have many advantages including suitable size, high stability, prolonged circulation time, elevated cargo capacity and controlled therapeutic release. Otherwise, the pathological features of some diseased cells, like cancer, allow PMs with particle size <200 nm to be passively uptaken via enhanced permeability and retention phenomenon (EPR). However, the passive targeting approach was proven to be insufficient in many cases. Consequently, the therapeutic efficiency of these PMs can be further reinforced by enhancing their cellular internalization via incorporating targeting ligands. These targeting ligands can enhance the assemblage of loaded cargos in the intended tissues via receptor-mediated endocytosis through exploiting receptors robustly expressed on the exterior of the intended tissue while minimizing their toxic effects. In this review, the up-to-date approaches of harnessing active targeting ligands to exploit certain overexpressed receptors will be summarized concerning the functionalization of the exterior of PMs for ameliorating their targeting potential in the scope of nanomedicine.
Collapse
Affiliation(s)
- Mona M Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre 33 El-Behooth St, Dokki Giza 12622 Egypt +202 33370931 +202 33371635
| | - Rehab Elsayed Marzouk
- Medical Biochemistry Department, Faculty of Medicine, Helwan University Helwan Cairo Egypt
| | - Sally A Sabra
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University Alexandria 21526 Egypt
| |
Collapse
|
25
|
Deng J, Yuan S, Pan W, Li Q, Chen Z. Nanotherapy to Reshape the Tumor Microenvironment: A New Strategy for Prostate Cancer Treatment. ACS OMEGA 2024; 9:26878-26899. [PMID: 38947792 PMCID: PMC11209918 DOI: 10.1021/acsomega.4c03055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024]
Abstract
Prostate cancer (PCa) is the second most common cancer in males worldwide. Androgen deprivation therapy (ADT) is the primary treatment method used for PCa. Although more effective androgen synthesis and antiandrogen inhibitors have been developed for clinical practice, hormone resistance increases the incidence of ADT-insensitive prostate cancer and poor prognoses. The tumor microenvironment (TME) has become a research hotspot with efforts to identify treatment targets based on the characteristics of the TME to improve prognosis. Herein, we introduce the basic characteristics of the PCa TME and the side effects of traditional prostate cancer treatments. We further highlight the emergence of novel nanotherapy strategies, their therapeutic mechanisms, and their effects on the PCa microenvironment. With further research, clinical applications of nanotherapy for PCa are expected in the near future. Collectively, this Review provides a valuable resource regarding the various nanotherapy types, demonstrating their broad clinical prospects to improve the quality of life in patients with PCa.
Collapse
Affiliation(s)
- Juan Deng
- The
Third Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325200, China
- The
First Clinical College of Guangdong Medical University, Zhanjiang, 524023, China
| | - Shaofei Yuan
- The
Third Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325200, China
| | - Wenjie Pan
- The
Third Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325200, China
| | - Qimeng Li
- The
Third Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325200, China
| | - Zhonglin Chen
- The
Third Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325200, China
| |
Collapse
|
26
|
Hou Y, Zhu C, Ban G, Shen Z, Liang Y, Chen K, Wang C, Shi H. Advancements and Challenges in the Application of Metal-Organic Framework (MOF) Nanocomposites for Tumor Diagnosis and Treatment. Int J Nanomedicine 2024; 19:6295-6317. [PMID: 38919774 PMCID: PMC11198007 DOI: 10.2147/ijn.s463144] [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: 02/06/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoscale metal-organic frameworks (MOFs) offer high biocompatibility, nanomaterial permeability, substantial specific surface area, and well-defined pores. These properties make MOFs valuable in biomedical applications, including biological targeting and drug delivery. They also play a critical role in tumor diagnosis and treatment, including tumor cell targeting, identification, imaging, and therapeutic methods such as drug delivery, photothermal effects, photodynamic therapy, and immunogenic cell death. The diversity of MOFs with different metal centers, organics, and surface modifications underscores their multifaceted contributions to tumor research and treatment. This review is a summary of these roles and mechanisms. The final section of this review summarizes the current state of the field and discusses prospects that may bring MOFs closer to pharmaceutical applications.
Collapse
Affiliation(s)
- Yingze Hou
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
- Clinical Medical College, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Can Zhu
- Department of Urology, The Second Clinical Medical College of Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Ge Ban
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Zhean Shen
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Yingbing Liang
- Department of Chemistry and Biotechnology, Graduate School of Engineering Tottori University Koyama-Minami 4-101, Tottori, 680-8552, Japan
| | - Kun Chen
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Chenbo Wang
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Heng Shi
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
27
|
Zheng H, An G, Yang X, Huang L, Wang N, Zhu Y. Iron-Based Metal-Organic Frameworks as Multiple Cascade Synergistic Therapeutic Effect Nano-Drug Delivery Systems for Effective Tumor Elimination. Pharmaceuticals (Basel) 2024; 17:812. [PMID: 38931479 PMCID: PMC11206809 DOI: 10.3390/ph17060812] [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: 05/16/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Efforts have been made to improve the therapeutic efficiency of tumor treatments, and metal-organic frameworks (MOFs) have shown excellent potential in tumor therapy. Monotherapy for the treatment of tumors has limited effects due to the limitation of response conditions and inevitable multidrug resistance, which seriously affect the clinical therapeutic effect. In this study, we chose to construct a multiple cascade synergistic tumor drug delivery system MIL-101(Fe)-DOX-TCPP-MnO2@PDA-Ag (MDTM@P-Ag) using MOFs as drug carriers. Under near-infrared (NIR) laser irradiation, 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) and Ag NPs loaded on MDTM@P-Ag can be activated to generate cytotoxic reactive oxygen species (ROS) and achieve photothermal conversion, thus effectively inducing the apoptosis of tumor cells and achieving a combined photodynamic/photothermal therapy. Once released at the tumor site, manganese dioxide (MnO2) can catalyze the decomposition of hydrogen peroxide (H2O2) in the acidic microenvironment of the tumor to generate oxygen (O2) and alleviate the hypoxic environment of the tumor. Fe3+/Mn2+ will mediate a Fenton/Fenton-like reaction to generate cytotoxic hydroxyl radicals (·OH), while depleting the high concentration of glutathione (GSH) in the tumor, thus enhancing the chemodynamic therapeutic effect. The successful preparation of the tumor drug delivery system and its good synergistic chemodynamic/photodynamic/photothermal therapeutic effect in tumor treatment can be demonstrated by the experimental results of material characterization, performance testing and in vitro experiments.
Collapse
Affiliation(s)
- Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Xiaohui Yang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Lei Huang
- School of Stomatology, Minzhu Clinic of Stomatology Hospital Affiliated to Guangxi Medical University, Nanning 530007, China;
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK
| |
Collapse
|
28
|
Wang Q, Yang Y, Li P, Dong R, Sun C, Song G, Wang Y. Titanium dioxide nanoparticles induce apoptosis through ROS-Ca 2+-p38/AKT/mTOR pathway in TM4 cells. J Appl Toxicol 2024; 44:818-832. [PMID: 38272789 DOI: 10.1002/jat.4583] [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: 10/25/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) can cause apoptosis in TM4 cells; however, the underlying mechanism has not been entirely elucidated. The purpose of this study was to investigate the effects of TiO2 NPs on ROS, Ca2+ level, p38/AKT/mTOR pathway, and apoptosis in TM4 cells and to evaluate the role of Ca2+ in p38/AKT/mTOR pathway and apoptosis. After exposure to different concentrations (0, 50, 100, 150, and 200 μg/mL) of TiO2 NPs for 24 h, cell viability, ROS, Ca2+ level, Ca2+-ATPase activity, p38/AKT/mTOR pathway-related proteins, apoptosis rate, and apoptosis-related proteins (Bax, Bcl-2, Caspase 3, Caspase 9, and p53) were detected. The ROS scavenger NAC was used to determine the effect of ROS on Ca2+ level. The Ca2+ chelator BAPTA-AM was used to evaluate the role of Ca2+ in p38/AKT/mTOR pathway and apoptosis. TiO2 NPs significantly inhibited cell viability, increased ROS level, and elevated Ca2+ level while suppressing Ca2+-ATPase activity. TiO2 NPs regulated the p38/AKT/mTOR pathway via increasing p-p38 level and decreasing p-AKT and p-mTOR levels. TiO2 NPs significantly enhanced the apoptosis. NAC attenuated Ca2+ overload and reduction in Ca2+-ATPase activity caused by TiO2 NPs. BAPTA-AM alleviated TiO2 NPs-induced abnormal expression of p38/AKT/mTOR pathway-related proteins. BAPTA-AM assuaged the apoptosis caused by TiO2 NPs. Altogether, this study revealed that TiO2 NPs elevated intracellular Ca2+ level through ROS accumulation. Subsequently, the heightened intracellular Ca2+ level was observed to exert regulation over the p38/AKT/mTOR pathway, ultimately culminating in apoptosis. These results provides a complementary understanding to the mechanism of TiO2 NPs-induced apoptosis in TM4 cells.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | | | - Pengfei Li
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Ruoyun Dong
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Chenhao Sun
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Guanling Song
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Yan Wang
- School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
29
|
Kar S, Das SS, Kundu S, Sahu BD, Kumar KJ, Kesari KK, Singh SK. Intranasal Delivery of Carvedilol- and Quercetin-Encapsulated Cationic Nanoliposomes for Cardiovascular Targeting: Formulation and In Vitro and Ex Vivo Studies. ACS APPLIED BIO MATERIALS 2024; 7:3061-3085. [PMID: 38581388 PMCID: PMC11530090 DOI: 10.1021/acsabm.4c00102] [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: 01/23/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024]
Abstract
Carvedilol (CVD), an adrenoreceptor blocker, is a hydrophobic Biopharmaceutics Classification System class II drug with poor oral bioavailability due to which frequent dosing is essential to attain pharmacological effects. Quercetin (QC), a polyphenolic compound, is a potent natural antioxidant, but its oral dosing is restricted due to poor aqueous solubility and low oral bioavailability. To overcome the common limitations of both drugs and to attain synergistic cardioprotective effects, we formulated CVD- and QC-encapsulated cationic nanoliposomes (NLPs) in situ gel (CVD/QC-L.O.F.) for intranasal administration. We designed CVD- and QC-loaded cationic nanoliposomal (NLPs) in situ gel (CVD/QC-L.O.F.) for intranasal administration. In vitro drug release studies of CVD/QC-L.O.F. (16.25%) exhibited 18.78 ± 0.57% of QC release and 91.38 ± 0.93% of CVD release for 120 h. Ex vivo nasal permeation studies of CVD/QC-L.O.F. demonstrated better permeation of QC (within 96 h), i.e., 75.09% compared to in vitro drug release, whereas CVD permeates within 48 h, indicating the better interaction between cationic NLPs and the negatively charged biological membrane. The developed nasal gel showed a sufficient mucoadhesive property, good spreadability, higher firmness, consistency, and cohesiveness, indicating suitability for membrane application and intranasal administration. CVD-NLPs, QC-NLPs, and CVD/QC-NLPs were evaluated for in vitro cytotoxicity, in vitro ROS-induced cell viability assessment, and a cellular uptake study using H9c2 rat cardiomyocytes. The highest in vitro cellular uptake of CVD/QC-cationic NLPs by H9c2 cells implies the benefit of QC loading within the CVD nanoliposomal carrier system and gives evidence for better interaction of NLPs carrying positive charges with the negatively charged biological cells. The in vitro H2O2-induced oxidative stress cell viability assessment of H9c2 cells established the intracellular antioxidant activity and cardioprotective effect of CVD/QC-cationic NLPs with low cytotoxicity. These findings suggest the potential of cationic NLPs as a suitable drug delivery carrier for CVD and QC combination for the intranasal route in the treatment of various cardiovascular diseases like hypertension, angina pectoris, etc. and for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Sweta Kar
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Sabya Sachi Das
- School
of Pharmaceutical and Population Health Informatics, DIT University, Dehradun 248009, Uttarakhand, India
| | - Sourav Kundu
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, Assam, India
| | - Bidya Dhar Sahu
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, Assam, India
| | - K. Jayaram Kumar
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Kavindra Kumar Kesari
- Department
of Applied Physics, School of Science, Aalto
University, 00076 Espoo, Finland
| | - Sandeep Kumar Singh
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| |
Collapse
|
30
|
Chen M, Qin Y, Peng Y, Mai R, Teng H, Qi Z, Mo J. Advancing stroke therapy: the potential of MOF-based nanozymes in biomedical applications. Front Bioeng Biotechnol 2024; 12:1363227. [PMID: 38798955 PMCID: PMC11119330 DOI: 10.3389/fbioe.2024.1363227] [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: 12/30/2023] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
In this study, we explored the growing use of metal-organic framework (MOF)-based Nanozymes in biomedical research, with a specific emphasis on their applications in stroke therapy. We have discussed the complex nature of stroke pathophysiology, highlighting the crucial role of reactive oxygen species (ROS), and acknowledging the limitations of natural enzymes in addressing these challenges. We have also discussed the role of nanozymes, particularly those based on MOFs, their structural similarities to natural enzymes, and their potential to improve reactivity in various biomedical applications. The categorization of MOF nanozymes based on enzyme-mimicking activities is discussed, and their applications in stroke therapy are explored. We have reported the potential of MOF in treating stroke by regulating ROS levels, alleviation inflammation, and reducing neuron apoptosis. Additionally, we have addressed the challenges in developing efficient antioxidant nanozyme systems for stroke treatment. The review concludes with the promise of addressing these challenges and highlights the promising future of MOF nanozymes in diverse medical applications, particularly in the field of stroke treatment.
Collapse
Affiliation(s)
- Meirong Chen
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Medical College of Guangxi University, Nanning, China
| | - Yang Qin
- Department of Graduate and Postgraduate Education Management, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yongmei Peng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Ruyu Mai
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Huanyao Teng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning, China
| | - Jingxin Mo
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Lab of Neurology, The Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
31
|
Shen J, Chen J, Qian Y, Wang X, Wang D, Pan H, Wang Y. Atomic Engineering of Single-Atom Nanozymes for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313406. [PMID: 38319004 DOI: 10.1002/adma.202313406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/24/2024] [Indexed: 02/07/2024]
Abstract
Single-atom nanozymes (SAzymes) showcase not only uniformly dispersed active sites but also meticulously engineered coordination structures. These intricate architectures bestow upon them an exceptional catalytic prowess, thereby captivating numerous minds and heralding a new era of possibilities in the biomedical landscape. Tuning the microstructure of SAzymes on the atomic scale is a key factor in designing targeted SAzymes with desirable functions. This review first discusses and summarizes three strategies for designing SAzymes and their impact on reactivity in biocatalysis. The effects of choices of carrier, different synthesis methods, coordination modulation of first/second shell, and the type and number of metal active centers on the enzyme-like catalytic activity are unraveled. Next, a first attempt is made to summarize the biological applications of SAzymes in tumor therapy, biosensing, antimicrobial, anti-inflammatory, and other biological applications from different mechanisms. Finally, how SAzymes are designed and regulated for further realization of diverse biological applications is reviewed and prospected. It is envisaged that the comprehensive review presented within this exegesis will furnish novel perspectives and profound revelations regarding the biomedical applications of SAzymes.
Collapse
Affiliation(s)
- Ji Shen
- Engineering Research Center of Advanced Rare Earth Materials, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jian Chen
- Institute of Science and Technology for New Energy, Xi'an Technological University, Xi'an, 710021, China
| | - Yuping Qian
- Center of Digital Dentistry/Department of Prosthodontics, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xinqiang Wang
- Institute of Science and Technology for New Energy, Xi'an Technological University, Xi'an, 710021, China
| | - Dingsheng Wang
- Engineering Research Center of Advanced Rare Earth Materials, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Hongge Pan
- Institute of Science and Technology for New Energy, Xi'an Technological University, Xi'an, 710021, China
| | - Yuguang Wang
- Center of Digital Dentistry/Department of Prosthodontics, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
32
|
Zheng Y, Chen X, Ding C, Liu X, Chi L, Zhang S. Abscisic acid ameliorates d-galactose -induced aging in mice by modulating AMPK-SIRT1-p53 pathway and intestinal flora. Heliyon 2024; 10:e28283. [PMID: 38524603 PMCID: PMC10957431 DOI: 10.1016/j.heliyon.2024.e28283] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024] Open
Abstract
Abscisic acid (ABA) is a plant hormone with various biological activities. Aging is a natural process accompanied by cognitive and physiological decline, and aging and its associated diseases pose a serious threat to public health, but its mechanisms remain insufficient. Therefore, the purpose of this study was to investigate the ameliorative effects of ABA on d-galactose (D-Gal)-induced aging in mice and to delve into its molecular mechanisms. Aging model was es-tablished by theintraperitoneal injection of D-Gal. We evaluated the oxidative stress by measuring superoxide dismutase (SOD), malondialdehyde (MDA), catalase (CAT) levels in serum. Proteins content in brain were determined by Western blot. D-Gal-induced brain damage was monitored by measuring the levels of acetylcholinesterase (AChE) content and hematoxylin-eosin staining (H&E). To evaluate the effects of ABA on aging, we measured the gut microbiota. The results demonstrated that ABA increased SOD, CAT and AChE, decreased MDA level. H&E staining showed that ABA could improve D-Gal-induced damage. In addition, ABA regulated the B-cell-lymphoma-2 (BCL-2) family and Phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) signaling pathway, while further regulating the acetylation of p53 protein by modulating the AMPK pathway and activating SIRT1 protein, thereby inhibiting the apoptosis of brain neurons and thus regulating the aging process. Interestingly, ABA improved the ratio of intestinal bacteria involved in regulating multiple metabolic pathways in the aging process, such as Bacteroides, Firmicutes, Lactobacillus and Ak-kermansia. In conclusion, the present study suggests that ABA may be responsible for improving and delaying the aging process by enhancing antioxidant activity, anti-apoptosis and regulating intestinal flora.
Collapse
Affiliation(s)
- Yongchun Zheng
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101, China
| | - Xueyan Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Chuanbo Ding
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101, China
- Changbai Mountain Characteristic Medicinal Resources Research and Development Innovation Center, Jilin, 132101, China
| | - Xinglong Liu
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101, China
- Changbai Mountain Characteristic Medicinal Resources Research and Development Innovation Center, Jilin, 132101, China
| | - Lihua Chi
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101, China
| | - Shuai Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
- Changbai Mountain Characteristic Medicinal Resources Research and Development Innovation Center, Jilin, 132101, China
| |
Collapse
|
33
|
Zhou Y, Li W. Methyltransferase-like 3-mediated m6A modification of miR-1908-5p contributes to nasopharyngeal carcinoma progression by targeting homeodomain-only protein homeobox. ENVIRONMENTAL TOXICOLOGY 2024; 39:1631-1640. [PMID: 38018881 DOI: 10.1002/tox.24032] [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: 08/07/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND N6-methyladenosine (m6A) modification interacting microRNAs (miRNAs) have been confirmed to participate in nasopharyngeal carcinoma (NPC) progression. This research investigated miR-1908-5p's function and regulatory mechanism in the tumorigenesis of NPC via m6A modification and targeting a key gene. METHODS The levels of miR-1908-5p, homeodomain-only protein homeobox (HOPX), and methyltransferase-like 3 (METTL3) expressions were detected via RT-qPCR. The correlation between miR-1908-5p and the HOPX/METTL3 axis, as well as their regulatory mechanism, was investigated by dual luciferase reporter, western blotting, and MeRIP assays. Moreover, the bio-functions of miR-1908-5p, HOPX, and METTL3 in NPC were explored through CCK8, transwell, caspase-3 activity, and xenograft tumor assays. RESULTS RT-qPCR results indicated a miR-1908-5p upregulation in NPC. Knocking down miR-1908-5p diminished the NPC cell viability and migration in vitro. In vivo, downregulating miR-1908-5p repressed NPC cell tumor growth. Moreover, HOPX was specifically targeted by miR-1908-5p, and HOPX downregulation led to reversal of the anti-tumor impact of the miR-1908-5p inhibitor against NPC cell malignancy. Also, METTL3 could mediate the m6A modification of miR-1908-5p to regulate its influence on NPC cells. CONCLUSION This study demonstrated that the METTL3-mediated m6A modification of miR-1908-5p enhanced the tumorigenesis of NPC by targeting HOPX. These findings propose new insights for NPC diagnosis and therapy.
Collapse
Affiliation(s)
- Yuanhong Zhou
- Department of Otolaryngology Head and Neck Surgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Wei Li
- Department of Otolaryngology Head and Neck Surgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
| |
Collapse
|
34
|
Li J, Geng H, Li X, Zou S, Xu X. RAD54B promotes gastric cancer cell migration and angiogenesis via the Wnt/β-catenin pathway. Radiol Oncol 2024; 58:67-77. [PMID: 38378037 PMCID: PMC10878776 DOI: 10.2478/raon-2024-0007] [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: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Gastric cancer is an epidemic malignancy that is commonly diagnosed at the late stage. Evidence has elucidated that RAD54B exerts a crucial role in the progress of various tumors, but its specific role and mechanism in gastric cancer remain gloomy. MATERIALS AND METHODS The level of RAD54B was detected by western blot. RAD54B expression was downregulated or upregulated in both MKN45 and AGS cells by the transfection of shRAD54B or overexpression plasmid, respectively. The role of RAD54B in the growth, migration, invasion and tube formation of gastric cancer was evaluated by Edu, colony formation, transwell and tube formation assays. In addition, the molecular mechanism of RAD54B in gastric cancer was also determined by western blot. Moreover, in vivo experiment was conducted in xenografted mice. RESULTS The expression of RAD54B was discovered to be upregulated in gastric cancer based on the ATGC and GEPIA databases, which was also confirmed in gastric cancer cell lines. Moreover, overexpression of RAD54B enhanced the growth, migration, invasion, tube formation and Wnt/β-catenin signaling axis in AGS and MKN45 cells. As expected, knockdown of RAD54B in AGS and MKN45 cells reversed these promotions. More importantly, in vivo assay also verified that RAD54B accelerated the growth of gastric cancer and Wnt/β-catenin signaling pathway. CONCLUSIONS Both loss-of-function and gain-of-function assays demonstrated that RAD54B facilitated gastric cancer cell progress and angiogenesis through the Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Jianchao Li
- Department of General Surgery, Changzhou TCM Hospital, Changzhou, Jiangsu, China
| | - Hui Geng
- Department of General Surgery, Changzhou TCM Hospital, Changzhou, Jiangsu, China
| | - Xin Li
- Department of General Surgery, Changzhou TCM Hospital, Changzhou, Jiangsu, China
| | - Shenshan Zou
- Department of General Surgery, Changzhou TCM Hospital, Changzhou, Jiangsu, China
| | - Xintao Xu
- Department of General Surgery, Changzhou TCM Hospital, Changzhou, Jiangsu, China
| |
Collapse
|
35
|
Gay MD, Drda JC, Chen W, Huang Y, Yassin AA, Duka T, Fang H, Shivapurkar N, Smith JP. Implicating the cholecystokinin B receptor in liver stem cell oncogenesis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G291-G309. [PMID: 38252699 PMCID: PMC11211039 DOI: 10.1152/ajpgi.00208.2023] [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: 09/25/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related deaths worldwide. Chronic inflammation and fibrosis are the greatest risk factors for the development of HCC. Although the cell of origin for HCC is uncertain, many theories believe this cancer may arise from liver progenitor cells or stem cells. Here, we describe the activation of hepatic stem cells that overexpress the cholecystokinin-B receptor (CCK-BR) after liver injury with either a DDC diet (0.1% 3, 5-diethoxy-carbonyl 1,4-dihydrocollidine) or a NASH-inducing CDE diet (choline-deficient ethionine) in murine models. Pharmacologic blockade of the CCK-BR with a receptor antagonist proglumide or knockout of the CCK-BR in genetically engineered mice during the injury diet reduces the expression of hepatic stem cells and prevents the formation of three-dimensional tumorspheres in culture. RNA sequencing of livers from DDC-fed mice treated with proglumide or DDC-fed CCK-BR knockout mice showed downregulation of differentially expressed genes involved in cell proliferation and oncogenesis and upregulation of tumor suppressor genes compared with controls. Inhibition of the CCK-BR decreases hepatic transaminases, fibrosis, cytokine expression, and alters the hepatic immune cell signature rendering the liver microenvironment less oncogenic. Furthermore, proglumide hastened recovery after liver injury by reversing fibrosis and improving markers of synthetic function. Proglumide is an older drug that is orally bioavailable and being repurposed for liver conditions. These findings support a promising therapeutic intervention applicable to patients to prevent the development of HCC and decrease hepatic fibrosis.NEW & NOTEWORTHY This investigation identified a novel pathway involving the activation of hepatic stem cells and liver oncogenesis. Receptor blockade or genetic disruption of the cholecystokinin-B receptor (CCK-BR) signaling pathway decreased the activation and proliferation of hepatic stem cells after liver injury without eliminating the regenerative capacity of healthy hepatocytes.
Collapse
Affiliation(s)
- Martha D Gay
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Jack C Drda
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Wenqiang Chen
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Yimeng Huang
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| | - Amal A Yassin
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| | - Tetyana Duka
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Hongbin Fang
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, District of Columbia, United States
| | - Narayan Shivapurkar
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Jill P Smith
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| |
Collapse
|
36
|
Ghosh A, Maske P, Patel V, Dubey J, Aniket K, Srivastava R. Theranostic applications of peptide-based nanoformulations for growth factor defective cancers. Int J Biol Macromol 2024; 260:129151. [PMID: 38181914 DOI: 10.1016/j.ijbiomac.2023.129151] [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: 07/01/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
Growth factors play a pivotal role in orchestrating cellular growth and division by binding to specific cell surface receptors. Dysregulation of growth factor production or activity can contribute to the uncontrolled cell proliferation observed in cancer. Peptide-based nanoformulations (PNFs) have emerged as promising therapeutic strategies for growth factor-deficient cancers. PNFs offer multifaceted capabilities including targeted delivery, imaging modalities, combination therapies, resistance modulation, and personalized medicine approaches. Nevertheless, several challenges remain, including limited specificity, stability, pharmacokinetics, tissue penetration, toxicity, and immunogenicity. To address these challenges and optimize PNFs for clinical translation, in-depth investigations are warranted. Future research should focus on elucidating the intricate interplay between peptides and nanoparticles, developing robust spectroscopic and computational methodologies, and establishing a comprehensive understanding of the structure-activity relationship governing peptide-nanoparticle interactions. Bridging these knowledge gaps will propel the translation of peptide-nanoparticle therapies from bench to bedside. While a few peptide-nanoparticle drugs have obtained FDA approval for cancer treatment, the integration of nanostructured platforms with peptide-based medications holds tremendous potential to expedite the implementation of innovative anticancer interventions. Therefore, growth factor-deficient cancers present both challenges and opportunities for targeted therapeutic interventions, with peptide-based nanoformulations positioned as a promising avenue. Nonetheless, concerted research and development endeavors are essential to optimize the specificity, stability, and safety profiles of PNFs, thereby advancing the field of peptide-based nanotherapeutics in the realm of oncology research.
Collapse
Affiliation(s)
- Arnab Ghosh
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| | - Priyanka Maske
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Vinay Patel
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Jyoti Dubey
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Kundu Aniket
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| | - Rohit Srivastava
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| |
Collapse
|
37
|
Wang W, Cheng Z, Xing H, Zhou S, Ye Q, Xiong G, Wang G, Ma D. Red cell membrane-coating Prussian blue for combined photothermal and NO gas therapy for nasopharyngeal carcinoma. J Mater Chem B 2024; 12:1579-1591. [PMID: 38259153 DOI: 10.1039/d3tb02444j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nitric oxide (NO) gas molecules have demonstrated remarkable anti-tumor effects and minimal susceptibility to drug resistance, establishing as a promising modality for effective tumor treatment. However, how to realize its stable and efficient delivery in vivo is still a challenge. In this study, we have developed a heat-responsive biomimetic nano erythrocyte (M/B@R) by loading a NO donor (BNN6) onto mesoporous Prussian blue (M-PB) and subsequently enveloping them with red blood cell membranes. The preserved integrity of the red blood cell membrane (RBCm) structure could ensure its excellent biosafety, prolong its circulation time within the bloodstream and then enhance the accumulation of BNN6 at tumor sites. When M/B@R is stimulated by near-infrared light (NIR-II, 808 nm) irradiation, the nanoparticle could generate significant heat for photothermal therapy (PTT) by the characteristic NIR absorption of M-PB and then NO could also be efficiently released. The generated NO further facilitates the formation of ONOO-, a highly toxic species to tumors, while also alleviating tumor hypoxia. Remarkably, M/B@R, with NIR as the excitation source, induces combined lethality through hyperthermia, DNA damage, and tumor hypoxia relief. This novel combination strategy provides a new avenue for PTT/NO-induced cancer therapy.
Collapse
Affiliation(s)
- Wenbo Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Zhaoyi Cheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Hui Xing
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Shihao Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Qiaozhang Ye
- Dalang Hospital of Dongguan, Dongguan 523000, China.
| | - Gaofei Xiong
- Dalang Hospital of Dongguan, Dongguan 523000, China.
| | - Guanhai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Dong Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
38
|
Shen Q, Yu C. Advances in superparamagnetic iron oxide nanoparticles modified with branched polyethyleneimine for multimodal imaging. Front Bioeng Biotechnol 2024; 11:1323316. [PMID: 38333548 PMCID: PMC10851169 DOI: 10.3389/fbioe.2023.1323316] [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: 10/17/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024] Open
Abstract
Multimodal imaging are approaches which combines multiple imaging techniques to obtain multi-aspect information of a target through different imaging modalities, thereby greatly improve the accuracy and comprehensiveness of imaging. Superparamagnetic iron oxide nanoparticles (SPIONs) modified with branched polyethyleneimine have revealed good biocompatibility and stability, high drug loading capacity and nucleic acid transfection efficiency. SPIONs have been developed as functionalized platforms which can be further modified to enhance their functionalities. Those further modifications facilitate the application of SPIONs in multimodal imaging. In this review, we discuss the methods, advantages, applications, and prospects of BPEI-modified SPIONs in multimodal imaging.
Collapse
Affiliation(s)
- Qiaoling Shen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
39
|
Sridhar A, Sekhon VK, Nguyen C, Abushalha K, Tahanan A, Rahbar MH, Jafri SH. Major Stressful Life Events and the Risk of Pancreatic, Head and Neck Cancers: A Case-Control Study. Cancers (Basel) 2024; 16:451. [PMID: 38275892 PMCID: PMC10814511 DOI: 10.3390/cancers16020451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Major stressful life events have been shown to be associated with an increased risk of lung cancer, breast cancer and the development of various chronic illnesses. The stress response generated by our body results in a variety of physiological and metabolic changes which can affect the immune system and have been shown to be associated with tumor progression. In this study, we aim to determine if major stressful life events are associated with the incidence of head and neck or pancreatic cancer (HNPC). METHODS This is a matched case-control study. Cases (CAs) were HNPC patients diagnosed within the previous 12 months. Controls (COs) were patients without a prior history of malignancy. Basic demographic data information on major stressful life events was collected using the modified Holmes-Rahe stress scale. A total sample of 280 was needed (79 cases, 201 controls) to achieve at least 80% power to detect odds ratios (ORs) of 2.00 or higher at the 5% level of significance. RESULTS From 1 January 2018 to 31 August 2021, 280 patients were enrolled (CA = 79, CO = 201) in this study. In a multivariable logistic regression analysis after controlling for potential confounding variables (including sex, age, race, education, marital status, smoking history), there was no difference between the lifetime prevalence of major stressful event in cases and controls. However, patients with HNPC were significantly more likely to report a major stressful life event within the preceding 5 years when compared to COs (p = 0.01, OR = 2.32, 95% CI, 1.18-4.54). CONCLUSIONS Patients with head, neck and pancreatic cancers are significantly associated with having a major stressful life event within 5 years of their diagnosis. This study highlights the potential need to recognize stressful life events as risk factors for developing malignancies.
Collapse
Affiliation(s)
- Arthi Sridhar
- Division of Hematology/Oncology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Vishaldeep Kaur Sekhon
- Division of Geriatric Medicine and Gerontology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chandler Nguyen
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Kamelah Abushalha
- MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA 01702, USA
| | - Amirali Tahanan
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (A.T.); (M.H.R.)
| | - Mohammad Hossein Rahbar
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (A.T.); (M.H.R.)
| | - Syed Hasan Jafri
- Division of Hematology/Oncology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
40
|
K R, S VK, Saravanan P, Rajeshkannan R, Rajasimman M, Kamyab H, Vasseghian Y. Exploring the diverse applications of Carbohydrate macromolecules in food, pharmaceutical, and environmental technologies. ENVIRONMENTAL RESEARCH 2024; 240:117521. [PMID: 37890825 DOI: 10.1016/j.envres.2023.117521] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/26/2023] [Accepted: 10/25/2023] [Indexed: 10/29/2023]
Abstract
Carbohydrates are a class of macromolecules that has significant potential across several domains, including the organisation of genetic material, provision of structural support, and facilitation of defence mechanisms against invasion. Their molecular diversity enables a vast array of essential functions, such as energy storage, immunological signalling, and the modification of food texture and consistency. Due to their rheological characteristics, solubility, sweetness, hygroscopicity, ability to prevent crystallization, flavour encapsulation, and coating capabilities, carbohydrates are useful in food products. Carbohydrates hold potential for the future of therapeutic development due to their important role in sustained drug release, drug targeting, immune antigens, and adjuvants. Bio-based packaging provides an emerging phase of materials that offer biodegradability and biocompatibility, serving as a substitute for traditional non-biodegradable polymers used as coatings on paper. Blending polyhydroxyalkanoates (PHA) with carbohydrate biopolymers, such as starch, cellulose, polylactic acid, etc., reduces the undesirable qualities of PHA, such as crystallinity and brittleness, and enhances the PHA's properties in addition to minimizing manufacturing costs. Carbohydrate-based biopolymeric nanoparticles are a viable and cost-effective way to boost agricultural yields, which is crucial for the increasing global population. The use of biopolymeric nanoparticles derived from carbohydrates is a potential and economically viable approach to enhance the quality and quantity of agricultural harvests, which is of utmost importance given the developing global population. The carbohydrate biopolymers may play in plant protection against pathogenic fungi by inhibiting spore germination and mycelial growth, may act as effective elicitors inducing the plant immune system to cope with pathogens. Furthermore, they can be utilised as carriers in controlled-release formulations of agrochemicals or other active ingredients, offering an alternative approach to conventional fungicides. It is expected that this review provides an extensive summary of the application of carbohydrates in the realms of food, pharmaceuticals, and environment.
Collapse
Affiliation(s)
- Ramaprabha K
- School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Venkat Kumar S
- Department of Petrochemical Technology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620 024, Tamil Nadu, India.
| | - Panchamoorthy Saravanan
- Department of Petrochemical Technology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620 024, Tamil Nadu, India
| | - R Rajeshkannan
- Department of Chemical Engineering, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| | - M Rajasimman
- Department of Chemical Engineering, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| | - Hesam Kamyab
- Faculty of Architecture and Urbanism, UTE University, Calle Rumipamba S/N and Bourgeois, Quito, Ecuador; Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600 077, India; Process Systems Engineering Centre (PROSPECT), Faculty of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Yasser Vasseghian
- Department of Chemistry, Soongsil University, Seoul, 06978, South Korea; School of Engineering, Lebanese American University, Byblos, Lebanon; University Centre for Research & Development, Department of Mechanical Engineering, Chandigarh University, Gharuan, Mohali, Punjab, 140413, India.
| |
Collapse
|
41
|
Meyers NL, Ashuach T, Lyons DE, Khalid MM, Simoneau CR, Erickson AL, Bouhaddou M, Nguyen TT, Kumar GR, Taha TY, Natarajan V, Baron JL, Neff N, Zanini F, Mahmoudi T, Quake SR, Krogan NJ, Cooper S, McDevitt TC, Yosef N, Ott M. Hepatitis C virus infects and perturbs liver stem cells. mBio 2023; 14:e0131823. [PMID: 37938000 PMCID: PMC10746249 DOI: 10.1128/mbio.01318-23] [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: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 11/09/2023] Open
Abstract
IMPORTANCE The hepatitis C virus (HCV) causes liver disease, affecting millions. Even though we have effective antivirals that cure HCV, they cannot stop terminal liver disease. We used an adult stem cell-derived liver organoid system to understand how HCV infection leads to the progression of terminal liver disease. Here, we show that HCV maintains low-grade infections in liver organoids for the first time. HCV infection in liver organoids leads to transcriptional reprogramming causing cancer cell development and altered immune response. Our finding shows how HCV infection in liver organoids mimics HCV infection and patient pathogenesis. These results reveal that HCV infection in liver organoids contributes to liver disease progression.
Collapse
Affiliation(s)
| | - Tal Ashuach
- Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California Berkeley, Berkeley, California, USA
| | | | - Mir M. Khalid
- Gladstone Institute of Virology, San Francisco, California, USA
| | | | - Ann L. Erickson
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Mehdi Bouhaddou
- Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California, San Francisco, California, USA
| | - Thong T. Nguyen
- Gladstone Institute of Virology, San Francisco, California, USA
| | - G. Renuka Kumar
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Taha Y. Taha
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Vaishaali Natarajan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Jody L. Baron
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Norma Neff
- Chan Zuckerburg Biohub, San Francisco, California, USA
| | - Fabio Zanini
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stephen R. Quake
- Chan Zuckerburg Biohub, San Francisco, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Nevan J. Krogan
- Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California, San Francisco, California, USA
| | - Stewart Cooper
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Todd C. McDevitt
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
- Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California Berkeley, Berkeley, California, USA
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Chan Zuckerburg Biohub, San Francisco, California, USA
| |
Collapse
|
42
|
Jain S, Malhotra KPK, Patiyal S, Raghava GPS. A Highly Accurate Model for Screening Prostate Cancer Using Propensity Index Panel of Ten Genes. J Comput Biol 2023; 30:1305-1314. [PMID: 37917795 DOI: 10.1089/cmb.2023.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Affiliation(s)
- Shipra Jain
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi, India
| | - Kawal Preet Kaur Malhotra
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi, India
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi, India
| | - Gajendra Pal Singh Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi, India
| |
Collapse
|
43
|
Lu Y, Wang S, Chi T, Zhao Y, Guo H, Wang H, Feng L. DNA damage repair-related gene signature for identifying the immune status and predicting the prognosis of hepatocellular carcinoma. Sci Rep 2023; 13:18978. [PMID: 37923899 PMCID: PMC10624694 DOI: 10.1038/s41598-023-45999-z] [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: 07/31/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
The heterogeneity of hepatocellular carcinoma (HCC) poses a challenge for accurate prognosis prediction. DNA damage repair genes (DDRGs) have an impact on a wide range of malignancies. However, the relevance of these genes in HCC prognosis has received little attention. In this study, we aimed to develop a prognostic signature to identify novel therapy options for HCC. We acquired mRNA expression profiles and clinical data for HCC patients from The Cancer Genome Atlas (TCGA) database. A polygenic prognostic model for HCC was constructed using selection operator Cox analysis and least absolute shrinkage. The model was validated using International Cancer Genome Consortium (ICGC) data. Overall survival (OS) between the high-risk and low-risk groups was compared using Kaplan‒Meier analysis. Independent predictors of OS were identified through both univariate and multivariate Cox analyses. To determine immune cell infiltration scores and activity in immune-related pathways, a single-sample gene set enrichment analysis was performed. The protein and mRNA expression levels of the prognostic genes between HCC and normal liver tissues were also examined by immunohistochemistry (IHC), immunofluorescence (IF) and quantitative real-time PCR (qRT-PCR). A novel ten-gene signature (CHD1L, HDAC1, KPNA2, MUTYH, PPP2R5B, NEIL3, POLR2L, RAD54B, RUVBL1 and SPP1) was established for HCC prognosis prediction. Patients in the high-risk group had worse OS than those in the low-risk group. Receiver operating characteristic curve analysis confirmed the predictive ability of this prognostic gene signature. Multivariate Cox analysis showed that the risk score was an independent predictor of OS. Functional analysis revealed a strong association with cell cycle and antigen binding pathways, and the risk score was highly correlated with tumor grade, tumor stage, and types of immune infiltrate. High expression levels of the prognostic genes were significantly correlated with increased sensitivity of cancer cells to antitumor drugs. IHC, IF and qRT-PCR all indicated that the prognostic genes were highly expressed in HCC relative to normal liver tissue, consistent with the results of bioinformatics analysis. Ten DDRGs were utilized to create a new signature for identifying the immunological state of HCC and predicting prognosis. In addition, blocking these genes could represent a promising treatment.
Collapse
Affiliation(s)
- Yongpan Lu
- Department of Plastic Surgery, Shandong University of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Jingshi Road, Jinan, 250014, Shandong, China
| | - Sen Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Tingting Chi
- Department of Acupuncture and Rehabilitation, The Affiliated Qingdao Hai Ci Hospital of Qingdao University (West Hospital Area), Qingdao, 266000, Shandong, China
| | - Yuli Zhao
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Huimin Guo
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Jining Medical College, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Haizheng Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China
| | - Li Feng
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qian Foshan Hospital, Shandong First Medical University, No. 16766, Jingshi Road, Jinan, 250014, Shandong, China.
| |
Collapse
|
44
|
Glassman I, Le N, Asif A, Goulding A, Alcantara CA, Vu A, Chorbajian A, Mirhosseini M, Singh M, Venketaraman V. The Role of Obesity in Breast Cancer Pathogenesis. Cells 2023; 12:2061. [PMID: 37626871 PMCID: PMC10453206 DOI: 10.3390/cells12162061] [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: 05/22/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Research has shown that obesity increases the risk for type 2 diabetes mellitus (Type 2 DM) by promoting insulin resistance, increases serum estrogen levels by the upregulation of aromatase, and promotes the release of reactive oxygen species (ROS) by macrophages. Increased circulating glucose has been shown to activate mammalian target of rapamycin (mTOR), a significant signaling pathway in breast cancer pathogenesis. Estrogen plays an instrumental role in estrogen-receptor-positive breast cancers. The role of ROS in breast cancer warrants continued investigation, in relation to both pathogenesis and treatment of breast cancer. We aim to review the role of obesity in breast cancer pathogenesis and novel therapies mediating obesity-associated breast cancer development. We explore the association between body mass index (BMI) and breast cancer incidence and the mechanisms by which oxidative stress modulates breast cancer pathogenesis. We discuss the role of glutathione, a ubiquitous antioxidant, in breast cancer therapy. Lastly, we review breast cancer therapies targeting mTOR signaling, leptin signaling, blood sugar reduction, and novel immunotherapy targets.
Collapse
Affiliation(s)
- Ira Glassman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Nghia Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Aamna Asif
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Anabel Goulding
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Cheldon Ann Alcantara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Annie Vu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Abraham Chorbajian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Mercedeh Mirhosseini
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Manpreet Singh
- Corona Regional Medical Center, Department of Emergency Medicine, Corona, CA 92882, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| |
Collapse
|
45
|
Pusta A, Tertis M, Crăciunescu I, Turcu R, Mirel S, Cristea C. Recent Advances in the Development of Drug Delivery Applications of Magnetic Nanomaterials. Pharmaceutics 2023; 15:1872. [PMID: 37514058 PMCID: PMC10383769 DOI: 10.3390/pharmaceutics15071872] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
With the predicted rise in the incidence of cancer, there is an ever-growing need for new cancer treatment strategies. Recently, magnetic nanoparticles have stood out as promising nanostructures for imaging and drug delivery systems as they possess unique properties. Moreover, magnetic nanomaterials functionalized with other compounds can lead to multicomponent nanoparticles with innovative structures and synergetic performance. The incorporation of chemotherapeutic drugs or RNA in magnetic drug delivery systems represents a promising alternative that can increase efficiency and reduce the side effects of anticancer therapy. This review presents a critical overview of the recent literature concerning the advancements in the field of magnetic nanoparticles used in drug delivery, with a focus on their classification, characteristics, synthesis and functionalization methods, limitations, and examples of magnetic drug delivery systems incorporating chemotherapeutics or RNA.
Collapse
Affiliation(s)
- Alexandra Pusta
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
- Department of Medical Devices, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Izabell Crăciunescu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Rodica Turcu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Simona Mirel
- Department of Medical Devices, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| |
Collapse
|
46
|
Chien ST, Suydam IT, Woodrow KA. Prodrug approaches for the development of a long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 198:114860. [PMID: 37160248 PMCID: PMC10498988 DOI: 10.1016/j.addr.2023.114860] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Long-acting formulations are designed to reduce dosing frequency and simplify dosing schedules by providing an extended duration of action. One approach to obtain long-acting formulations is to combine long-acting prodrugs (LA-prodrug) with existing or emerging drug delivery technologies (DDS). The design criteria for long-acting prodrugs are distinct from conventional prodrug strategies that alter absorption, distribution, metabolism, and excretion (ADME) parameters. Our review focuses on long-acting prodrug delivery systems (LA-prodrug DDS), which is a subcategory of long-acting formulations where prodrug design enables DDS formulation to achieve an extended duration of action that is greater than the parent drug. Here, we define LA-prodrugs as the conjugation of an active pharmaceutical ingredient (API) to a promoiety group via a cleavable covalent linker, where both the promoiety and linker are selected to enable formulation and administration from a drug delivery system (DDS) to achieve an extended duration of action. These LA-prodrug DDS results in an extended interval where the API is within a therapeutic range without necessarily altering ADME as is typical of conventional prodrugs. The conversion of the LA-prodrug to the API is dependent on linker cleavage, which can occur before or after release from the DDS. The requirement for linker cleavage provides an additional tool to prolong release from these LA-prodrug DDS. In addition, the physicochemical properties of drugs can be tuned by promoiety selection for a particular DDS. Conjugation with promoieties that are carriers or amenable to assembly into carriers can also provide access to formulations designed for extending duration of action. LA-prodrugs have been applied to a wide variety of drug delivery strategies and are categorized in this review by promoiety size and complexity. Small molecule promoieties (typically MW < 1000 Da) have been used to improve encapsulation or partitioning as well as broaden APIs for use with traditional long-acting formulations such as solid drug dispersions. Macromolecular promoieties (typically MW > 1000 Da) have been applied to hydrogels, nanoparticles, micelles, dendrimers, and polymerized prodrug monomers. The resulting LA-prodrug DDS enable extended duration of action for active pharmaceuticals across a wide range of applications, with target release timescales spanning days to years.
Collapse
Affiliation(s)
- Shin-Tian Chien
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Ian T Suydam
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
47
|
Sharma A, Kaur I, Dheer D, Nagpal M, Kumar P, Venkatesh DN, Puri V, Singh I. A propitious role of marine sourced polysaccharides: Drug delivery and biomedical applications. Carbohydr Polym 2023; 308:120448. [PMID: 36813329 DOI: 10.1016/j.carbpol.2022.120448] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Numerous compounds, with extensive applications in biomedical and biotechnological fields, are present in the oceans, which serve as a prime renewable source of natural substances, further promoting the development of novel medical systems and devices. Polysaccharides are present in the marine ecosystem in abundance, promoting minimal extraction costs, in addition to their solubility in extraction media, and an aqueous solvent, along with their interactions with biological compounds. Certain algae-derived polysaccharides include fucoidan, alginate, and carrageenan, while animal-derived polysaccharides comprise hyaluronan, chitosan and many others. Furthermore, these compounds can be modified to facilitate their processing into multiple shapes and sizes, as well as exhibit response dependence to external conditions like temperature and pH. All these properties have promoted the use of these biomaterials as raw materials for the development of drug delivery carrier systems (hydrogels, particles, capsules). The present review enlightens marine polysaccharides providing its sources, structures, biological properties, and its biomedical applications. In addition to this, their role as nanomaterials is also portrayed by the authors, along with the methods employed to develop them and associated biological and physicochemical properties designed to develop suitable drug delivery systems.
Collapse
Affiliation(s)
- Ameya Sharma
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; University of Glasgow, College of Medical, Veterinary and Life Sciences, Glasgow, United Kingdom, G12 8QQ
| | - Divya Dheer
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D Nagasamy Venkatesh
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Vivek Puri
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India.
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
48
|
Yang Z, Xie H, Wan J, Wang Y, Zhang L, Zhou K, Tang H, Zhao W, Wang H, Song P, Zheng S. A nanotherapeutic strategy that engages cytotoxic and immunosuppressive activities for the treatment of cancer recurrence following organ transplantation. EBioMedicine 2023; 92:104594. [PMID: 37167784 DOI: 10.1016/j.ebiom.2023.104594] [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: 10/19/2022] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Long-term treatment with immunosuppressants is necessary to attenuate allograft rejection following organ transplantation (OT). Consequently, the overall survival of OT recipients with malignancies has been substantially compromised by tumour recurrence. Rapamycin (RAPA) is a clinically approved immunosuppressive agent with antitumour activity that is considered beneficial in preventing posttransplant tumour recurrence. However, the clinical outcome of RAPA is impeded by acquired drug resistance and its poor oral bioavailability. METHODS A nanotherapeutic strategy was developed by supramolecular assembly of RAPA into a polymer cytotoxic 7-ethyl-10-hydroxycamptothecin (SN38) prodrug nanoparticle (termed SRNP) for simultaneous codelivery of cytotoxic/immunosuppressive agents. Cell-based experiments were used to evaluate the cytotoxicity of SRNPs against hepatocellular carcinoma (HCC). The therapeutic efficacy of SRNPs was evaluated in multiple preclinical models including an orthotopic HCC mouse model, an orthotopic liver transplantation (OLT) rat model and a clinically relevant cancer-transplant model to examine its antitumour and immunosuppressive activity. FINDINGS The combination of SN38 with RAPA resulted in synergetic effects against HCC cells and alleviated RAPA resistance by abrogating Akt/mTOR signalling activation. SRNPs exhibited potent antitumour efficiency in the orthotopic HCC model while substantially prolonging the survival of allografts in the OLT model. In the cancer-transplant model that simultaneously bears tumour xenografts and skin allografts, SRNPs not only effectively inhibited tumour growth but also attenuated allograft damage. INTERPRETATION The nanotherapy presented here had enhanced efficacy against tumours and maintained satisfactory immunosuppressive activity and thus has great potential to improve the survival outcomes of patients with a high risk of tumour recurrence following OT. FUNDING This work was supported by the National Natural Science Foundation of China (32171368 and 31671019), the Zhejiang Provincial Natural Science Foundation of China (LZ21H180001), the Zhejiang Province Preeminence Youth Fund (LR19H160002), and the Jinan Provincial Laboratory Research Project of Microecological Biomedicine (JNL-2022039c).
Collapse
Affiliation(s)
- Zhentao Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Jianqin Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuchen Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Liang Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ke Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hong Tang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wentao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hangxiang Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou 310003, Zhejiang Province, China.
| |
Collapse
|
49
|
Nairuz T, Mahmud Z, Manik RK, Kabir Y. Cancer stem cells: an insight into the development of metastatic tumors and therapy resistance. Stem Cell Rev Rep 2023:10.1007/s12015-023-10529-x. [PMID: 37129728 DOI: 10.1007/s12015-023-10529-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 05/03/2023]
Abstract
The term "cancer stem cells" (CSCs) refers to cancer cells that exhibit traits parallel to normal stem cells, namely the potential to give rise to every type of cell identified in a tumor microenvironment. It has been found that CSCs usually develops from other neoplastic cells or non-cancerous somatic cells by acquiring stemness and malignant characteristics through particular genetic modifications. A trivial number of CSCs, identified in solid and liquid cancer, can give rise to an entire tumor population with aggressive anticancer drug resistance, metastasis, and invasiveness. Besides, cancer stem cells manipulate their intrinsic and extrinsic features, regulate the metabolic pattern of the cell, adjust efflux-influx efficiency, modulate different signaling pathways, block apoptotic signals, and cause genetic and epigenetic alterations to retain their pluripotency and ability of self-renewal. Notably, to keep the cancer stem cells' ability to become malignant cells, mesenchymal stem cells, tumor-associated fibroblasts, immune cells, etc., interact with one another. Furthermore, CSCs are characterized by the expression of particular molecular markers that carry significant diagnostic and prognostic significance. Because of this, scientific research on CSCs is becoming increasingly imperative, intending to understand the traits and behavior of cancer stem cells and create more potent anticancer therapeutics to fight cancer at the CSC level. In this review, we aimed to elucidate the critical role of CSCs in the onset and spread of cancer and the characteristics of CSCs that promote severe resistance to targeted therapy.
Collapse
Affiliation(s)
- Tahsin Nairuz
- Department of Biochemistry and Molecular Biology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Rasel Khan Manik
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
50
|
Li H, Zhuang H, Gu T, Li G, Jiang Y, Xu S, Zhou Q. RAD54L promotes progression of hepatocellular carcinoma via the homologous recombination repair pathway. Funct Integr Genomics 2023; 23:128. [PMID: 37071224 DOI: 10.1007/s10142-023-01060-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high incidence worldwide. The underlying mechanisms remain poorly understood. The DNA metabolic process of homologous recombination repair (HRR) has been linked to a high probability of tumorigenesis and drug resistance. This study aimed to determine the role of HRR in HCC and identify critical HRR-related genes that affect tumorigenesis and prognosis. A total of 613 tumor and 252 para-carcinoma tissue samples were collected from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) to obtain differentially expressed genes (DEGs). HRR-related genes were assessed using gene enrichment and pathway analyses. Survival analysis was performed using the Kaplan-Meier method in the Gene Expression Profiling Interactive Analysis portal. The levels of RAD54L in the HRR pathway were detected by RT-qPCR and western blotting in para-carcinoma and HCC tissues and in L02 normal human liver cells and Huh7 HCC cells. Immunohistochemistry (IHC) was performed on the clinical specimens to determine the connection between gene expression and clinical features. Bioinformatics analysis revealed that the HRR pathway was enriched in HCC tissues. Upregulation of HRR pathway DEGs in HCC tissues was positively correlated with tumor pathological staging and negatively associated with patient overall survival. RAD54B, RAD54L, and EME1 genes in the HRR pathway were screened as markers for predicting HCC prognosis. RT-qPCR identified RAD54L as the most significantly expressed of the three genes. Western blotting and IHC quantitative analyses further demonstrated that RAD54L protein levels were higher in HCC tissues. IHC analysis of 39 pairs of HCC and para-carcinoma tissue samples also revealed an association between RAD54L and Edmondson-Steiner grade and the proliferation-related gene Ki67. The collective findings positively correlate RAD54L in the HRR signaling pathway with HCC staging and implicate RAD54L as a marker to predict HCC progression.
Collapse
Affiliation(s)
- Hongda Li
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Haiwen Zhuang
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| | - Tengfei Gu
- Department of Anesthesiology, People's Hospital of Lianshui County, Huai'an, China
| | - Guangyu Li
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuhang Jiang
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Sanrong Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qing Zhou
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|