Pub Date : 2024-10-12eCollection Date: 2024-01-01DOI: 10.2147/IJN.S478930
Aftab Ahmed Khan, Abdulaziz Abdullah AlKhureif, Manal Almutairi, Abrar Nasser Bin Nooh, Saeed Awod Bin Hassan, Yasser M Alqahtani
Introduction: There is a critical need to address concerns surrounding the potential impact of bleaching gels specifically on the tooth substrate. Therefore, this laboratory investigation aimed to assess the impact of a hydrogen peroxide (HP)-free bleaching (HiSmileTM) in comparison to an HP-based bleaching (Opalescence RegularTM) on the surface and mechanical characteristics of tooth substrate.
Methods: Sixty sound human premolar teeth were sectioned to produce dentin fragments and divided into two primary groups based on the bleaching agent used. Each group was subdivided into three subgroups (n = 10) per distinct bleaching regimens: (T1) fragments underwent a 7-day immersion in distilled water at 37°C without any bleaching treatment, (T2) fragments underwent a 7-day immersion in distilled water at 37°C, with the application of bleaching gel occurring on the seventh day for 10 minutes, and (T3) fragments underwent a bleaching regimen for seven consecutive days, each session lasting for 10 minutes. The initial and final evaluations of surface roughness, nano-hardness, and elastic modulus were performed. Following the bleaching regimens of T3, a composite stub was fabricated on the dentin fragments for the shear bond strength (SBS) test. Statistical testing was accomplished using the analysis of variance (P < 0.05).
Results: HP-based bleaching gel showed significant differences between measurement intervals in surface roughness, elastic modulus, and SBS parameters (P < 0.05). In contrast, HP-free bleaching gel showed insignificant differences within the group (P > 0.05). The SBS between dentin-composite was significantly affected with the use of HP-based bleaching gel, while HP-free bleaching gel showed insignificant difference between measurement intervals. The qualitative validation of the treatment's impact was further demonstrated using the scanning electron microscopy.
Conclusion: The findings suggest that the bonding stability of composite restorations to dentin may be compromised after bleaching with an HP-based gel, whereas immediate bonding procedures can be safely conducted following the application of an HP-free bleaching gel.
简介:目前亟需解决漂白凝胶对牙齿基质的潜在影响问题。因此,本实验室调查旨在评估不含过氧化氢(HP)的漂白剂(HiSmileTM)与含HP的漂白剂(Opalescence RegularTM)相比对牙齿基质表面和机械特性的影响:对 60 颗健全的人类前臼齿进行切片,制作牙本质碎片,并根据所使用的漂白剂将其分为两组。每组又按不同的漂白方案分为三个亚组(n = 10):(T1)牙片在 37°C 的蒸馏水中浸泡 7 天,不进行任何漂白处理;(T2)牙片在 37°C 的蒸馏水中浸泡 7 天,第七天涂漂白凝胶,每次 10 分钟;(T3)牙片连续漂白 7 天,每次 10 分钟。对表面粗糙度、纳米硬度和弹性模量进行了初步和最终评估。T3 漂白疗程结束后,在牙本质碎片上制作复合材料桩,进行剪切粘接强度(SBS)测试。统计检验采用方差分析法(P < 0.05):结果:基于 HP 的漂白凝胶在表面粗糙度、弹性模量和 SBS 参数方面显示出不同测量区间的显著差异(P < 0.05)。相比之下,不含 HP 的漂白凝胶在组内差异不显著(P > 0.05)。使用基于 HP 的漂白凝胶后,牙本质-复合材料之间的 SBS 受到明显影响,而不含 HP 的漂白凝胶在不同测量间隔之间的差异不明显。结论:研究结果表明,牙本质与复合材料之间的 SBS 在使用 HP 漂白凝胶后会受到明显影响,而不含 HP 漂白凝胶的牙本质与复合材料之间的 SBS 在不同测量时间段内差异不明显:结论:研究结果表明,使用含HP的漂白凝胶进行漂白后,复合树脂修复体与牙本质的粘接稳定性可能会受到影响,而使用不含HP的漂白凝胶则可以安全地进行即刻粘接。
{"title":"Effects of Time-Elapsed Bleaching on the Surface and Mechanical Properties of Dentin Substrate Using Hydrogen Peroxide-Free Nanohydroxyapatite Gel.","authors":"Aftab Ahmed Khan, Abdulaziz Abdullah AlKhureif, Manal Almutairi, Abrar Nasser Bin Nooh, Saeed Awod Bin Hassan, Yasser M Alqahtani","doi":"10.2147/IJN.S478930","DOIUrl":"https://doi.org/10.2147/IJN.S478930","url":null,"abstract":"<p><strong>Introduction: </strong>There is a critical need to address concerns surrounding the potential impact of bleaching gels specifically on the tooth substrate. Therefore, this laboratory investigation aimed to assess the impact of a hydrogen peroxide (HP)-free bleaching (HiSmile<sup>TM</sup>) in comparison to an HP-based bleaching (Opalescence Regular<sup>TM</sup>) on the surface and mechanical characteristics of tooth substrate.</p><p><strong>Methods: </strong>Sixty sound human premolar teeth were sectioned to produce dentin fragments and divided into two primary groups based on the bleaching agent used. Each group was subdivided into three subgroups (n = 10) per distinct bleaching regimens: (T<sub>1</sub>) fragments underwent a 7-day immersion in distilled water at 37°C without any bleaching treatment, (T<sub>2</sub>) fragments underwent a 7-day immersion in distilled water at 37°C, with the application of bleaching gel occurring on the seventh day for 10 minutes, and (T<sub>3</sub>) fragments underwent a bleaching regimen for seven consecutive days, each session lasting for 10 minutes. The initial and final evaluations of surface roughness, nano-hardness, and elastic modulus were performed. Following the bleaching regimens of T<sub>3</sub>, a composite stub was fabricated on the dentin fragments for the shear bond strength (SBS) test. Statistical testing was accomplished using the analysis of variance (P < 0.05).</p><p><strong>Results: </strong>HP-based bleaching gel showed significant differences between measurement intervals in surface roughness, elastic modulus, and SBS parameters (P < 0.05). In contrast, HP-free bleaching gel showed insignificant differences within the group (P > 0.05). The SBS between dentin-composite was significantly affected with the use of HP-based bleaching gel, while HP-free bleaching gel showed insignificant difference between measurement intervals. The qualitative validation of the treatment's impact was further demonstrated using the scanning electron microscopy.</p><p><strong>Conclusion: </strong>The findings suggest that the bonding stability of composite restorations to dentin may be compromised after bleaching with an HP-based gel, whereas immediate bonding procedures can be safely conducted following the application of an HP-free bleaching gel.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10307-10317"},"PeriodicalIF":6.6,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11485028/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-12eCollection Date: 2024-01-01DOI: 10.2147/IJN.S476667
Tarek A Ahmed, Ghada A Milibary, Alshaimaa M Almehmady, Amerh A Alahmadi, Ehab M M Ali, Khalid M El-Say
Background: This study investigates the influence of various formulation parameters on the characteristics of hinokitiol-loaded phytosomes and evaluates their anticancer potential against breast cancer cells.
Materials and methods: Phytosomal nanoparticles were prepared and characterized for size, zeta potential, and entrapment efficiency. Morphological analysis was conducted using optical microscopy and transmission electron microscopy (TEM). The solubility of hinokitiol at different pH levels was determined, and the in vitro release profile of the optimized phytosomes was assessed. Cytotoxicity assays were performed to evaluate the anticancer efficacy against breast cancer cell lines, and apoptosis induction was examined using Annexin V/propidium iodide staining. Cell cycle analysis was conducted to assess the impact on cell cycle progression.
Results: The optimized phytosomes demonstrated a size range of 138.4 ± 7.7 to 763.7 ± 15.4 nm, with zeta potentials ranging from -10.2 ± 0.28 to -53.2 ± 1.06 mV and entrapment efficiencies between 29.161 ± 1.163% and 92.77 ± 7.01%. Morphological characterization confirmed uniformity and spherical morphology. Hinokitiol solubility increased with pH, and the release from the optimized phytosomes exhibited sustained patterns. The formulated phytosomes showed superior cytotoxicity, with lower IC50 values compared to pure hinokitiol. Treatment induced significant apoptosis and cell cycle arrest at the G2/M and S phases.
Conclusion: Hinokitiol-loaded phytosomes demonstrate promising anticancer efficacy against breast cancer cells, highlighting their potential as targeted therapeutic agents for breast cancer therapy.
{"title":"Improving the Cytotoxic Activity of Hinokitiol from Drug-Loaded Phytosomal Formulation Against Breast Cancer Cell Lines.","authors":"Tarek A Ahmed, Ghada A Milibary, Alshaimaa M Almehmady, Amerh A Alahmadi, Ehab M M Ali, Khalid M El-Say","doi":"10.2147/IJN.S476667","DOIUrl":"https://doi.org/10.2147/IJN.S476667","url":null,"abstract":"<p><strong>Background: </strong>This study investigates the influence of various formulation parameters on the characteristics of hinokitiol-loaded phytosomes and evaluates their anticancer potential against breast cancer cells.</p><p><strong>Materials and methods: </strong>Phytosomal nanoparticles were prepared and characterized for size, zeta potential, and entrapment efficiency. Morphological analysis was conducted using optical microscopy and transmission electron microscopy (TEM). The solubility of hinokitiol at different pH levels was determined, and the in vitro release profile of the optimized phytosomes was assessed. Cytotoxicity assays were performed to evaluate the anticancer efficacy against breast cancer cell lines, and apoptosis induction was examined using Annexin V/propidium iodide staining. Cell cycle analysis was conducted to assess the impact on cell cycle progression.</p><p><strong>Results: </strong>The optimized phytosomes demonstrated a size range of 138.4 ± 7.7 to 763.7 ± 15.4 nm, with zeta potentials ranging from -10.2 ± 0.28 to -53.2 ± 1.06 mV and entrapment efficiencies between 29.161 ± 1.163% and 92.77 ± 7.01%. Morphological characterization confirmed uniformity and spherical morphology. Hinokitiol solubility increased with pH, and the release from the optimized phytosomes exhibited sustained patterns. The formulated phytosomes showed superior cytotoxicity, with lower IC50 values compared to pure hinokitiol. Treatment induced significant apoptosis and cell cycle arrest at the G2/M and S phases.</p><p><strong>Conclusion: </strong>Hinokitiol-loaded phytosomes demonstrate promising anticancer efficacy against breast cancer cells, highlighting their potential as targeted therapeutic agents for breast cancer therapy.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10321-10339"},"PeriodicalIF":6.6,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481998/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464715","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-12eCollection Date: 2024-01-01DOI: 10.2147/IJN.S496595
Jiale Zhang
{"title":"Nanoparticle-Mediated Delivery of Natural Anti-Inflammatories for Cardiovascular Disease [Letter].","authors":"Jiale Zhang","doi":"10.2147/IJN.S496595","DOIUrl":"https://doi.org/10.2147/IJN.S496595","url":null,"abstract":"","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10319-10320"},"PeriodicalIF":6.6,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481988/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464633","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-11eCollection Date: 2024-01-01DOI: 10.2147/IJN.S448876
Shouye Sun, Qian Wang, Bin Zhang, Yutao Cui, Xinghui Si, Gan Wang, Jingwei Wang, Hang Xu, Baoming Yuan, Chuangang Peng
Purpose: During treatment of infected bone defects, control of infection is necessary for effective bone repair, and hence controlled topical application of antibiotics is required in clinical practice. In this study, a biodegradable drug delivery system with in situ gelation at the site of infection was prepared by integrating vancomycin into a polyethylene glycol/oxidized dextran (PEG/ODEX) hydrogel matrix.
Methods: In this work, PEG/ODEX hydrogels were prepared by Schiff base reaction, and vancomycin was loaded into them to construct a drug delivery system with controllable release and degradability. We first examined the microstructure, degradation time and drug release of the hydrogels. Then we verified the biocompatibility and in vitro ability of the release system. Finally, we used a rat infected bone defect model for further experiments.
Results: The results showed that this antibacterial system could be completely biodegradable in vivo for 56 days, and its degradation products did not cause specific inflammatory response. The cumulative release of vancomycin from the antibacterial system was 58.3% ± 3.8% at 14 days and 78.4% ± 3.2% at 35 days. The concentration of vancomycin in the surrounding environment was about 1.2 mg/mL, which can effectively remove bacteria. Further studies in vivo showed that the antibacterial system cleared the infection and accelerated repair of infected bone defects in the femur of rats. There was no infection in rats after 8 weeks of treatment. The 3D image analysis of the experimental group showed that the bone volume fraction (BV/TV) was 1.39-fold higher (p < 0.001), the trabecular number (Tb.N) was 1.31-fold higher (p < 0.05), and the trabecular separation (Tb.Sp) was 0.58-fold higher than those of the control group (p < 0.01).
Conclusion: In summary, this study clearly demonstrates that a clinical strategy based on biological materials can provide an innovative and effective approach to treatment of infected bone defects.
{"title":"Vancomycin-Loaded in situ Gelled Hydrogel as an Antibacterial System for Enhancing Repair of Infected Bone Defects.","authors":"Shouye Sun, Qian Wang, Bin Zhang, Yutao Cui, Xinghui Si, Gan Wang, Jingwei Wang, Hang Xu, Baoming Yuan, Chuangang Peng","doi":"10.2147/IJN.S448876","DOIUrl":"https://doi.org/10.2147/IJN.S448876","url":null,"abstract":"<p><strong>Purpose: </strong>During treatment of infected bone defects, control of infection is necessary for effective bone repair, and hence controlled topical application of antibiotics is required in clinical practice. In this study, a biodegradable drug delivery system with in situ gelation at the site of infection was prepared by integrating vancomycin into a polyethylene glycol/oxidized dextran (PEG/ODEX) hydrogel matrix.</p><p><strong>Methods: </strong>In this work, PEG/ODEX hydrogels were prepared by Schiff base reaction, and vancomycin was loaded into them to construct a drug delivery system with controllable release and degradability. We first examined the microstructure, degradation time and drug release of the hydrogels. Then we verified the biocompatibility and in vitro ability of the release system. Finally, we used a rat infected bone defect model for further experiments.</p><p><strong>Results: </strong>The results showed that this antibacterial system could be completely biodegradable in vivo for 56 days, and its degradation products did not cause specific inflammatory response. The cumulative release of vancomycin from the antibacterial system was 58.3% ± 3.8% at 14 days and 78.4% ± 3.2% at 35 days. The concentration of vancomycin in the surrounding environment was about 1.2 mg/mL, which can effectively remove bacteria. Further studies in vivo showed that the antibacterial system cleared the infection and accelerated repair of infected bone defects in the femur of rats. There was no infection in rats after 8 weeks of treatment. The 3D image analysis of the experimental group showed that the bone volume fraction (BV/TV) was 1.39-fold higher (<i>p</i> < 0.001), the trabecular number (Tb.N) was 1.31-fold higher (<i>p</i> < 0.05), and the trabecular separation (Tb.Sp) was 0.58-fold higher than those of the control group (<i>p</i> < 0.01).</p><p><strong>Conclusion: </strong>In summary, this study clearly demonstrates that a clinical strategy based on biological materials can provide an innovative and effective approach to treatment of infected bone defects.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10227-10245"},"PeriodicalIF":6.6,"publicationDate":"2024-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11476785/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-10eCollection Date: 2024-01-01DOI: 10.2147/IJN.S480639
Shuqi Gao, Zhe Sui, Qian Jiang, Yueyao Jiang
Niosomes are key nanocarriers composed of bilayer vesicles formed by non-ionic surfactants and cholesterol, offering advantages such as high physicochemical stability, biodegradability, cost-effectiveness, and low toxicity. This review discusses their significant role in drug delivery, including applications in anticancer therapy and vaccine delivery. It also highlights the impact of non-ionic surfactants on niosome formation, drug delivery pathways, and protein corona formation-a relatively underexplored topic. Furthermore, the application of artificial intelligence in optimizing niosome design and functionality is examined. Future research directions include enhancing formulation techniques, expanding application scopes, and integrating advanced technologies. This review provides comprehensive insights and practical guidance for advancing niosome-based drug delivery systems.
{"title":"Functional Evaluation of Niosomes Utilizing Surfactants in Nanomedicine Applications.","authors":"Shuqi Gao, Zhe Sui, Qian Jiang, Yueyao Jiang","doi":"10.2147/IJN.S480639","DOIUrl":"https://doi.org/10.2147/IJN.S480639","url":null,"abstract":"<p><p>Niosomes are key nanocarriers composed of bilayer vesicles formed by non-ionic surfactants and cholesterol, offering advantages such as high physicochemical stability, biodegradability, cost-effectiveness, and low toxicity. This review discusses their significant role in drug delivery, including applications in anticancer therapy and vaccine delivery. It also highlights the impact of non-ionic surfactants on niosome formation, drug delivery pathways, and protein corona formation-a relatively underexplored topic. Furthermore, the application of artificial intelligence in optimizing niosome design and functionality is examined. Future research directions include enhancing formulation techniques, expanding application scopes, and integrating advanced technologies. This review provides comprehensive insights and practical guidance for advancing niosome-based drug delivery systems.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10283-10305"},"PeriodicalIF":6.6,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11472738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Breast cancer treatment has been a global puzzle, and targeted strategies based on the hypoxic tumor microenvironment (TME) have attracted extensive attention. As a signature transcription factor overexpressed in hypoxia tumor, hypoxia-inducible factor-1 (HIF-1) contribute to cancer progression. Compound 7-(3-(2-chloro-1H-benzo[d]1midazole-1-yl) propoxy)-2-(3,4,5-trime-thoxyphenyl)-4H-chromen-4-one, synthesized and named FB15 in our earlier research, a potential inhibitor of HIF-1α signaling pathway, has been proved a promising drug candidate for many kinds of cancer chemotherapy. However, the poor solubility and undesirable pharmacokinetics of FB15 leads to limited treatment efficacy of tumor, which ultimately restricts its potential clinical applications. Carbonic anhydrase IX (CAIX), a tumor cell transmembrane protein, was overexpressed in hypoxia tumor site. Acetazolamide (AZA), a highly selective ligand targeting CAIX, can be utilized to delivery FB15 to hypoxia tumor site.
Methods: In this study, we prepared and characterized FB15 loaded nano-mixed micelles with the AZA conjugated poloxamer 188 (AZA-P188) and D-a-Tocopherol Polyethylene 1000 Glycol Succinate (TPGS), denoted as, AZA-P188/TPGS@FB15. Its delivery efficiency in vitro and in vivo was assessed by in vitro drug release, cytotoxicity assay, cellular uptake, and in vivo pharmacokinetics and fluorescence imaging. Finally, therapeutic effect of AZA-P188/TPGS@FB15 was investigated using a preclinical breast cancer subcutaneous graft model in vivo.
Results: In vitro studies revealed that AZA-P188/TPGS@FB15 could efficiently target breast cancer cells mediated by CAIX receptor, trigger FB15 release in response to acidic condition, and enhance cellular uptake and cytotoxicity against breast cancer cells. The pharmacokinetic studies showed that FB15-loaded AZA-functionalized micelles exhibited significantly increased AUC0-t over free FB15. In vivo imaging demonstrated that AZA-functionalized micelles significantly increased the drug distribution in the tumor site. In vivo experiments confirmed that AZA-P188/TPGS@FB15 exhibited superior inhibition of tumor growth in nude mice with good biosafety.
Conclusion: AZA-P188/TPGS@FB15 hold promise as a potentially effective therapeutic way for breast cancer. Its targeted delivery system utilizing AZA as a carrier shows potential for improving the efficacy of FB15 in cancer therapy.
{"title":"A Smart CA IX-Targeting and pH-Responsive Nano-Mixed Micelles for Delivery of FB15 with Superior Anti-Breast Cancer Efficacy.","authors":"XingYun Liu, JingDuo Zhao, FuRong Liu, ZhiZhong Xie, XiaoYong Lei, Zhe Wang, Zerui Yang, YuSheng Zhou, GuoTao Tang","doi":"10.2147/IJN.S459047","DOIUrl":"https://doi.org/10.2147/IJN.S459047","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer treatment has been a global puzzle, and targeted strategies based on the hypoxic tumor microenvironment (TME) have attracted extensive attention. As a signature transcription factor overexpressed in hypoxia tumor, hypoxia-inducible factor-1 (HIF-1) contribute to cancer progression. Compound 7-(3-(2-chloro-1H-benzo[d]1midazole-1-yl) propoxy)-2-(3,4,5-trime-thoxyphenyl)-4H-chromen-4-one, synthesized and named FB15 in our earlier research, a potential inhibitor of HIF-1α signaling pathway, has been proved a promising drug candidate for many kinds of cancer chemotherapy. However, the poor solubility and undesirable pharmacokinetics of FB15 leads to limited treatment efficacy of tumor, which ultimately restricts its potential clinical applications. Carbonic anhydrase IX (CAIX), a tumor cell transmembrane protein, was overexpressed in hypoxia tumor site. Acetazolamide (AZA), a highly selective ligand targeting CAIX, can be utilized to delivery FB15 to hypoxia tumor site.</p><p><strong>Methods: </strong>In this study, we prepared and characterized FB15 loaded nano-mixed micelles with the AZA conjugated poloxamer 188 (AZA-P188) and D-a-Tocopherol Polyethylene 1000 Glycol Succinate (TPGS), denoted as, AZA-P188/TPGS@FB15. Its delivery efficiency in vitro and in vivo was assessed by in vitro drug release, cytotoxicity assay, cellular uptake, and in vivo pharmacokinetics and fluorescence imaging. Finally, therapeutic effect of AZA-P188/TPGS@FB15 was investigated using a preclinical breast cancer subcutaneous graft model in vivo.</p><p><strong>Results: </strong>In vitro studies revealed that AZA-P188/TPGS@FB15 could efficiently target breast cancer cells mediated by CAIX receptor, trigger FB15 release in response to acidic condition, and enhance cellular uptake and cytotoxicity against breast cancer cells. The pharmacokinetic studies showed that FB15-loaded AZA-functionalized micelles exhibited significantly increased AUC<sub>0-t</sub> over free FB15. In vivo imaging demonstrated that AZA-functionalized micelles significantly increased the drug distribution in the tumor site. In vivo experiments confirmed that AZA-P188/TPGS@FB15 exhibited superior inhibition of tumor growth in nude mice with good biosafety.</p><p><strong>Conclusion: </strong>AZA-P188/TPGS@FB15 hold promise as a potentially effective therapeutic way for breast cancer. Its targeted delivery system utilizing AZA as a carrier shows potential for improving the efficacy of FB15 in cancer therapy.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10247-10262"},"PeriodicalIF":6.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11471892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-09eCollection Date: 2024-01-01DOI: 10.2147/IJN.S467968
Xiaojun Zhang, Ying Zhao, Zhaogang Teng, Tangyao Sun, Jun Tao, Jiang Wu, Yu Wang, Fan Qiu, Feng Wang
Introduction: Photothermal therapy (PTT) is a promising therapeutic procedure with minimal side effects, which can not only kill tumor directly but also cause immunogenic cell death (ICD). However, most solid tumors, including neuroblastoma, are abundant in fibroblasts, which limit the penetration and delivery of nanoparticles. Losartan is an antihypertensive drug approved by the FDA, and it has been proved to have the effect of breaking down excessive ECM network.
Methods: In this study, we investigated the application and potential mechanism of the combination of mesoporous platinum nanoparticles (MPNs) and losartan in the PTT of neuroblastoma by establishing neuroblastoma models in vitro and in vivo.
Results: Compared to the MPNs group without 808 nm laser irradiation, Neuro-2a cells pretreated with PTT and losartan showed lower survival rates, increased surface calreticulin, and higher release of HMGB1 and ATP. The group also exhibited the highest anti-tumor efficacy in vivo, with a tumor suppression ratio of approximately 80%. Meanwhile, we found that CD3+ T cells, CD4+ T cells and CD8+ T cells from the peripheral blood of experimental group mice were significantly higher than control groups, and CD8+PD-1+ cells were significantly lower than those in MPNs + Los group and Los + laser group. And the expression of PD-1 and α-SMA in Neuro-2a tumors tissue was reduced. Furthermore, losartan could reduce damage of liver function caused by MPNs and laser treatment.
Conclusion: This study demonstrated that losartan-induced fibroblasts ablation increased the penetration of MPNs into tumors. Enhanced penetration allowed PTT to kill more tumor cells and synergistically activate immune cells, leading to ICD, indicating the great promise of the strategy for treating neuroblastoma in vivo.
{"title":"Combination of Losartan and Platinum Nanoparticles with Photothermal Therapy Induces Immunogenic Cell Death Effective Against Neuroblastoma.","authors":"Xiaojun Zhang, Ying Zhao, Zhaogang Teng, Tangyao Sun, Jun Tao, Jiang Wu, Yu Wang, Fan Qiu, Feng Wang","doi":"10.2147/IJN.S467968","DOIUrl":"https://doi.org/10.2147/IJN.S467968","url":null,"abstract":"<p><strong>Introduction: </strong>Photothermal therapy (PTT) is a promising therapeutic procedure with minimal side effects, which can not only kill tumor directly but also cause immunogenic cell death (ICD). However, most solid tumors, including neuroblastoma, are abundant in fibroblasts, which limit the penetration and delivery of nanoparticles. Losartan is an antihypertensive drug approved by the FDA, and it has been proved to have the effect of breaking down excessive ECM network.</p><p><strong>Methods: </strong>In this study, we investigated the application and potential mechanism of the combination of mesoporous platinum nanoparticles (MPNs) and losartan in the PTT of neuroblastoma by establishing neuroblastoma models in vitro and in vivo.</p><p><strong>Results: </strong>Compared to the MPNs group without 808 nm laser irradiation, Neuro-2a cells pretreated with PTT and losartan showed lower survival rates, increased surface calreticulin, and higher release of HMGB1 and ATP. The group also exhibited the highest anti-tumor efficacy in vivo, with a tumor suppression ratio of approximately 80%. Meanwhile, we found that CD3<sup>+</sup> T cells, CD4<sup>+</sup> T cells and CD8<sup>+</sup> T cells from the peripheral blood of experimental group mice were significantly higher than control groups, and CD8<sup>+</sup>PD-1<sup>+</sup> cells were significantly lower than those in MPNs + Los group and Los + laser group. And the expression of PD-1 and α-SMA in Neuro-2a tumors tissue was reduced. Furthermore, losartan could reduce damage of liver function caused by MPNs and laser treatment.</p><p><strong>Conclusion: </strong>This study demonstrated that losartan-induced fibroblasts ablation increased the penetration of MPNs into tumors. Enhanced penetration allowed PTT to kill more tumor cells and synergistically activate immune cells, leading to ICD, indicating the great promise of the strategy for treating neuroblastoma in vivo.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10213-10226"},"PeriodicalIF":6.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11471086/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-09eCollection Date: 2024-01-01DOI: 10.2147/IJN.S479068
Khaled S Allemailem, Ahmad Almatroudi, Faris Alrumaihi, Arwa Essa Alradhi, Abdulrahman Theyab, Mohammad Algahtani, Mohmmed Othman Alhawas, Gasim Dobie, Amira A Moawad, Arshad Husain Rahmani, Amjad Ali Khan
The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated sequence (CRISPR/Cas) system is a cutting-edge genome-editing tool employed to explore the functions of normal and disease-related genes. The CRISPR/Cas system has a remarkable diversity in the composition and architecture of genomic loci and Cas protein sequences. Owing to its excellent efficiency and specificity, this system adds an outstanding dimension to biomedical research on genetic manipulation of eukaryotic cells. However, safe, efficient, and specific delivery of this system to target cells and tissues and their off-target effects are considered critical bottlenecks for the therapeutic applications. Recently discovered anti-CRISPR proteins (Acr) play a significant role in limiting the effects of this system. Acrs are relatively small proteins that are highly specific to CRISPR variants and exhibit remarkable structural diversity. The in silico approaches, crystallography, and cryo-electron microscopy play significant roles in elucidating the mechanisms of action of Acrs. Acrs block the CRISPR/Cas system mainly by employing four mechanisms: CRISPR/Cas complex assembly interruption, target-binding interference, target cleavage prevention, and degradation of cyclic oligonucleotide signaling molecules. Engineered CRISPR/Cas systems are frequently used in gene therapy, diagnostics, and functional genomics. Understanding the molecular mechanisms underlying Acr action may help in the safe and effective use of CRISPR/Cas tools for genetic modification, particularly in the context of medicine. Thus, attempts to regulate prokaryotic CRISPR/Cas surveillance complexes will advance the development of antimicrobial drugs and treatment of human diseases. In this review, recent updates on CRISPR/Cas systems, especially CRISPR/Cas9 and Acrs, and their novel mechanistic insights are elaborated. In addition, the role of Acrs in the novel applications of CRISPP/Cas biotechnology for precise genome editing and other applications is discussed.
{"title":"Current Updates of CRISPR/Cas System and Anti-CRISPR Proteins: Innovative Applications to Improve the Genome Editing Strategies.","authors":"Khaled S Allemailem, Ahmad Almatroudi, Faris Alrumaihi, Arwa Essa Alradhi, Abdulrahman Theyab, Mohammad Algahtani, Mohmmed Othman Alhawas, Gasim Dobie, Amira A Moawad, Arshad Husain Rahmani, Amjad Ali Khan","doi":"10.2147/IJN.S479068","DOIUrl":"https://doi.org/10.2147/IJN.S479068","url":null,"abstract":"<p><p>The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated sequence (CRISPR/Cas) system is a cutting-edge genome-editing tool employed to explore the functions of normal and disease-related genes. The CRISPR/Cas system has a remarkable diversity in the composition and architecture of genomic loci and Cas protein sequences. Owing to its excellent efficiency and specificity, this system adds an outstanding dimension to biomedical research on genetic manipulation of eukaryotic cells. However, safe, efficient, and specific delivery of this system to target cells and tissues and their off-target effects are considered critical bottlenecks for the therapeutic applications. Recently discovered anti-CRISPR proteins (Acr) play a significant role in limiting the effects of this system. Acrs are relatively small proteins that are highly specific to CRISPR variants and exhibit remarkable structural diversity. The in silico approaches, crystallography, and cryo-electron microscopy play significant roles in elucidating the mechanisms of action of Acrs. Acrs block the CRISPR/Cas system mainly by employing four mechanisms: CRISPR/Cas complex assembly interruption, target-binding interference, target cleavage prevention, and degradation of cyclic oligonucleotide signaling molecules. Engineered CRISPR/Cas systems are frequently used in gene therapy, diagnostics, and functional genomics. Understanding the molecular mechanisms underlying Acr action may help in the safe and effective use of CRISPR/Cas tools for genetic modification, particularly in the context of medicine. Thus, attempts to regulate prokaryotic CRISPR/Cas surveillance complexes will advance the development of antimicrobial drugs and treatment of human diseases. In this review, recent updates on CRISPR/Cas systems, especially CRISPR/Cas9 and Acrs, and their novel mechanistic insights are elaborated. In addition, the role of Acrs in the novel applications of CRISPP/Cas biotechnology for precise genome editing and other applications is discussed.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10185-10212"},"PeriodicalIF":6.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11471075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464707","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-09eCollection Date: 2024-01-01DOI: 10.2147/IJN.S475320
Ye Ming, Xinyi He, Zhenxing Zhao, Xuehuan Meng, Ye Zhu, Hao Tan, Guoyin Yang, Yun Hu, Leilei Zheng
Purpose: Oxidative stress and mitochondrial dysfunction are potential contributors to the compromised tissue regeneration capacity of alveolar bone in diabetic patients. Berberine, an active plant alkaloid, exhibits multiple pharmacological effects including antioxidation, blood glucose- and blood lipid-lowering properties. However, it remains uncertain whether berberine can improve impaired osteogenesis in type 2 diabetes mellitus (T2DM), and its poor solubility and oral bioavailability also constrain its applications in bone regeneration. Thus, our study aimed to probe the effects of berberine on bone marrow stem cells (BMSCs) in a diabetic microenvironment, with a greater emphasis on developing a suitable nano-delivery system for berberine and assessing its capability to repair diabetic alveolar bone defects.
Methods: Firstly, BMSCs were exposed to berberine within a high glucose and palmitate (HG+PA) environment. Reactive oxygen species levels, mitochondrial membrane potential, ATP generation, cell apoptosis, and osteogenic potential were subsequently assessed. Next, we explored the regulatory mechanism of autophagy flux in the positive effects of berberine. Furthermore, a nanocarrier based on emulsion electrospinning for sustained local delivery of berberine (Ber@SF/PCL) was established. We assessed its capacity to enhance bone healing in the alveolar bone defect of T2DM rats through micro-computed tomography and histology analysis.
Results: Berberine treatment could inhibit reactive oxygen species overproduction, mitochondrial dysfunction, apoptosis, and improve osteogenesis differentiation by restoring autophagy flux under HG+PA conditions. Notably, Ber@SF/PCL electrospun nanofibrous membrane with excellent physicochemical properties and good biological safety had the potential to promote alveolar bone remodeling in T2DM rats.
Conclusion: Our study shed new lights into the protective role of berberine on BMSCs under T2DM microenvironment. Furthermore, berberine-loaded composite electrospun membrane may serve as a promising approach for regenerating alveolar bone in diabetic patients.
{"title":"Nanocarrier-Assisted Delivery of Berberine Promotes Diabetic Alveolar Bone Regeneration by Scavenging ROS and Improving Mitochondrial Dysfunction.","authors":"Ye Ming, Xinyi He, Zhenxing Zhao, Xuehuan Meng, Ye Zhu, Hao Tan, Guoyin Yang, Yun Hu, Leilei Zheng","doi":"10.2147/IJN.S475320","DOIUrl":"https://doi.org/10.2147/IJN.S475320","url":null,"abstract":"<p><strong>Purpose: </strong>Oxidative stress and mitochondrial dysfunction are potential contributors to the compromised tissue regeneration capacity of alveolar bone in diabetic patients. Berberine, an active plant alkaloid, exhibits multiple pharmacological effects including antioxidation, blood glucose- and blood lipid-lowering properties. However, it remains uncertain whether berberine can improve impaired osteogenesis in type 2 diabetes mellitus (T2DM), and its poor solubility and oral bioavailability also constrain its applications in bone regeneration. Thus, our study aimed to probe the effects of berberine on bone marrow stem cells (BMSCs) in a diabetic microenvironment, with a greater emphasis on developing a suitable nano-delivery system for berberine and assessing its capability to repair diabetic alveolar bone defects.</p><p><strong>Methods: </strong>Firstly, BMSCs were exposed to berberine within a high glucose and palmitate (HG+PA) environment. Reactive oxygen species levels, mitochondrial membrane potential, ATP generation, cell apoptosis, and osteogenic potential were subsequently assessed. Next, we explored the regulatory mechanism of autophagy flux in the positive effects of berberine. Furthermore, a nanocarrier based on emulsion electrospinning for sustained local delivery of berberine (Ber@SF/PCL) was established. We assessed its capacity to enhance bone healing in the alveolar bone defect of T2DM rats through micro-computed tomography and histology analysis.</p><p><strong>Results: </strong>Berberine treatment could inhibit reactive oxygen species overproduction, mitochondrial dysfunction, apoptosis, and improve osteogenesis differentiation by restoring autophagy flux under HG+PA conditions. Notably, Ber@SF/PCL electrospun nanofibrous membrane with excellent physicochemical properties and good biological safety had the potential to promote alveolar bone remodeling in T2DM rats.</p><p><strong>Conclusion: </strong>Our study shed new lights into the protective role of berberine on BMSCs under T2DM microenvironment. Furthermore, berberine-loaded composite electrospun membrane may serve as a promising approach for regenerating alveolar bone in diabetic patients.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10263-10282"},"PeriodicalIF":6.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11471107/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464632","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-07eCollection Date: 2024-01-01DOI: 10.2147/IJN.S479848
Hui Peng, Qian Jiang, Wenhao Mao, Zhonglan Hu, Qi Wang, Zhuo Yu, Li Zhang, Xinyan Wang, Chunbo Zhuang, Jia Mai, Zhiyuan Wang, Ting Sun
Background: The hypoxic tumor microenvironment and single mechanisms severely limit the photodynamic therapy (PDT) efficiency of covalent organic framework (COF) nanoparticles in cancer treatment.
Purpose: Here, we propose an iron-loaded, hydrophilic 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000)-modified hollow covalent organic framework (HCOF), Fe-HCOF-PEG2000, for use in hypoxic PDT and ferroptosis therapy owing to its type I and II photodynamic ability and iron nanoparticle loading property.
Results: Fe-HCOF-PEG2000 nanoparticles (Fe-HCOFs-PEG2000) with semiconducting polymers and microporous skeletons allow efficient photophysical properties. Moreover, the iron nanoparticles on Fe-HCOF-PEG2000 caused ferroptosis and further enhanced tumor elimination under normoxic and hypoxic conditions. DSPE-PEG2000 endowed Fe-HCOF-PEG2000 with hydrophilicity, allowing it to circulate and accumulate in organs rich in blood supply, especially tumors. 808 nm NIR activated Fe-HCOF-PEG2000 aggregated in tumors and significantly inhibited tumor growth under hypoxia.
Conclusion: To our knowledge, Fe-HCOF-PEG2000 is the leading combination of type I/II PDT and ferroptosis. The strong antitumor effects of this nanomaterial suggest prospects for clinical translation as a tumor nanotherapy drug.
{"title":"Fe-HCOF-PEG<sup>2000</sup> as a Hypoxia-Tolerant Photosensitizer to Trigger Ferroptosis and Enhance ROS-Based Cancer Therapy.","authors":"Hui Peng, Qian Jiang, Wenhao Mao, Zhonglan Hu, Qi Wang, Zhuo Yu, Li Zhang, Xinyan Wang, Chunbo Zhuang, Jia Mai, Zhiyuan Wang, Ting Sun","doi":"10.2147/IJN.S479848","DOIUrl":"https://doi.org/10.2147/IJN.S479848","url":null,"abstract":"<p><strong>Background: </strong>The hypoxic tumor microenvironment and single mechanisms severely limit the photodynamic therapy (PDT) efficiency of covalent organic framework (COF) nanoparticles in cancer treatment.</p><p><strong>Purpose: </strong>Here, we propose an iron-loaded, hydrophilic 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000)-modified hollow covalent organic framework (HCOF), Fe-HCOF-PEG<sup>2000</sup>, for use in hypoxic PDT and ferroptosis therapy owing to its type I and II photodynamic ability and iron nanoparticle loading property.</p><p><strong>Results: </strong>Fe-HCOF-PEG<sup>2000</sup> nanoparticles (Fe-HCOFs-PEG<sup>2000</sup>) with semiconducting polymers and microporous skeletons allow efficient photophysical properties. Moreover, the iron nanoparticles on Fe-HCOF-PEG<sup>2000</sup> caused ferroptosis and further enhanced tumor elimination under normoxic and hypoxic conditions. DSPE-PEG<sup>2000</sup> endowed Fe-HCOF-PEG<sup>2000</sup> with hydrophilicity, allowing it to circulate and accumulate in organs rich in blood supply, especially tumors. 808 nm NIR activated Fe-HCOF-PEG<sup>2000</sup> aggregated in tumors and significantly inhibited tumor growth under hypoxia.</p><p><strong>Conclusion: </strong>To our knowledge, Fe-HCOF-PEG<sup>2000</sup> is the leading combination of type I/II PDT and ferroptosis. The strong antitumor effects of this nanomaterial suggest prospects for clinical translation as a tumor nanotherapy drug.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"19 ","pages":"10165-10183"},"PeriodicalIF":6.6,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11468433/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}