Pub Date : 2024-09-27eCollection Date: 2024-01-01DOI: 10.2147/IJN.S480722
Ruien Chen, Huamei Zou, Xiuwen Ye, Bailing Xie, Aizhi Zhang, Lihua Mo, Yu Liu, Huanping Zhang, Gui Yang, Pingchang Yang
Introduction: The therapeutic efficacy for airway allergies needs to be improved. Th2 polarization is a primary pathological feature of airway allergies. We constructed chimeric antigen-LgDNA (Lactobacillus rhamnosus DNA) nanoparticles (CAP-NPs). The effects of CAP-NPs on reconciling airway Th2 polarization were tested.
Methods: In this study, disulfide bond-linked antigen-major histocompatibility complex II (MHC II)-LgDNA nanoparticles (NPs) were constructed and designated CAP-NPs. An airway Th2 polarization mouse model was established to test the effects of CAP-NPs on suppressing the Th2 response.
Results: The CAP-NP components of ovalbumin (OVA), major histocompatibility complex II (MHC II), and LgDNA were confirmed in a series of laboratory tests. The CAP-NPs remained stable at pH7.2 for at least 96 h. In in vitro experiments, CAP-NPs bound to the surface of OVA-specific CD4+ T cells, which resulted in apoptosis of the antigen-specific CD4+ T cells. Removal of any of the three components from the NPs abolished the induction of apoptosis of antigen specific CD4+ T cells. CAP-NPs increased the expression of lysine-specific demethylase 5A (KDM5A) in CD4+ T cells. Histone H3K9 and the gene promoter of caspase 8 were demethylated by KDM5A, which led to transcription and expression of the caspase 8 gene. Administration of CAP-NPs significantly alleviated experimental airway Th2 polarization through activating the caspase 8-apoptosis signaling pathway.
Discussion: In this paper, we constructed CAP-NPs that could induce antigen-specific CD4+ T cell apoptosis. Administration of CAP-NPs efficiently alleviated experimental airway Th2 polarization.
简介气道过敏的治疗效果有待提高。Th2 极化是气道过敏的主要病理特征。我们构建了嵌合抗原-LgDNA(鼠李糖乳杆菌 DNA)纳米颗粒(CAP-NPs)。测试了 CAP-NPs 对调和气道 Th2 极化的影响:本研究构建了二硫键连接的抗原-主要组织相容性复合体 II(MHC II)-LgDNA 纳米颗粒(NPs),并将其命名为 CAP-NPs。建立了气道Th2极化小鼠模型,以检验CAP-NPs抑制Th2反应的效果:结果:一系列实验室测试证实,CAP-NP的成分包括卵清蛋白(OVA)、主要组织相容性复合体II(MHC II)和LgDNA。在体外实验中,CAP-NPs 与 OVA 特异性 CD4+ T 细胞表面结合,导致抗原特异性 CD4+ T 细胞凋亡。去除 NPs 中的三种成分中的任何一种都不会诱导抗原特异性 CD4+ T 细胞凋亡。CAP-NPs 增加了 CD4+ T 细胞中赖氨酸特异性去甲基化酶 5A (KDM5A) 的表达。组蛋白 H3K9 和 caspase 8 基因启动子被 KDM5A 去甲基化,从而导致 caspase 8 基因的转录和表达。通过激活caspase 8-凋亡信号通路,服用CAP-NPs可明显缓解实验性气道Th2极化:本文构建了可诱导抗原特异性 CD4+ T 细胞凋亡的 CAP-NPs。服用CAP-NPs能有效缓解实验性气道Th2极化。
{"title":"Chimeric Antigen-LgDNA Nanoparticles Attenuate Airway Th2 Polarization.","authors":"Ruien Chen, Huamei Zou, Xiuwen Ye, Bailing Xie, Aizhi Zhang, Lihua Mo, Yu Liu, Huanping Zhang, Gui Yang, Pingchang Yang","doi":"10.2147/IJN.S480722","DOIUrl":"10.2147/IJN.S480722","url":null,"abstract":"<p><strong>Introduction: </strong>The therapeutic efficacy for airway allergies needs to be improved. Th2 polarization is a primary pathological feature of airway allergies. We constructed chimeric antigen-LgDNA (<i>Lactobacillus rhamnosus</i> DNA) nanoparticles (CAP-NPs). The effects of CAP-NPs on reconciling airway Th2 polarization were tested.</p><p><strong>Methods: </strong>In this study, disulfide bond-linked antigen-major histocompatibility complex II (MHC II)-LgDNA nanoparticles (NPs) were constructed and designated CAP-NPs. An airway Th2 polarization mouse model was established to test the effects of CAP-NPs on suppressing the Th2 response.</p><p><strong>Results: </strong>The CAP-NP components of ovalbumin (OVA), major histocompatibility complex II (MHC II), and LgDNA were confirmed in a series of laboratory tests. The CAP-NPs remained stable at pH7.2 for at least 96 h. In in vitro experiments, CAP-NPs bound to the surface of OVA-specific CD4<sup>+</sup> T cells, which resulted in apoptosis of the antigen-specific CD4<sup>+</sup> T cells. Removal of any of the three components from the NPs abolished the induction of apoptosis of antigen specific CD4<sup>+</sup> T cells. CAP-NPs increased the expression of lysine-specific demethylase 5A (KDM5A) in CD4<sup>+</sup> T cells. Histone H3K9 and the gene promoter of caspase 8 were demethylated by KDM5A, which led to transcription and expression of the caspase 8 gene. Administration of CAP-NPs significantly alleviated experimental airway Th2 polarization through activating the caspase 8-apoptosis signaling pathway.</p><p><strong>Discussion: </strong>In this paper, we constructed CAP-NPs that could induce antigen-specific CD4<sup>+</sup> T cell apoptosis. Administration of CAP-NPs efficiently alleviated experimental airway Th2 polarization.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11444059/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142361476","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}
Introduction: Alzheimer's disease (AD), a neurodegenerative condition, stands as the most prevalent form of dementia. Its complex pathological mechanisms and the formidable blood-brain barrier (BBB) pose significant challenges to current treatment approaches. Oxidative stress is recognized as a central factor in AD, underscoring the importance of antioxidative strategies in its treatment. In this study, we developed a novel brain-targeted nanoparticle, Ce/Zr-MOF@Cur-Lf, for AD therapy.
Methods: Layer-by-layer self-assembly technology was used to prepare Ce/Zr-MOF@Cur-Lf. In addition, the effect on the intracellular reactive oxygen species level, the uptake effect by PC12 and bEnd.3 cells and the in vitro BBB permeation effect were investigated. Finally, the mouse AD model was established by intrahippocampal injection of Aβ1-42, and the in vivo biodistribution, AD therapeutic effect and biosafety of the nanoparticles were researched at the animal level.
Results: As anticipated, Ce/Zr-MOF@Cur-Lf demonstrated efficient BBB penetration and uptake by PC12 cells, leading to attenuation of H2O2-induced oxidative damage. Moreover, intravenous administration of Ce/Zr-MOF@Cur-Lf resulted in rapid brain access and improvement of various pathological features of AD, including neuronal damage, amyloid-β deposition, dysregulated central cholinergic system, oxidative stress, and neuroinflammation.
Conclusion: Overall, Ce/Zr-MOF@Cur-Lf represents a promising approach for precise brain targeting and multi-target mechanisms in AD therapy, potentially serving as a viable option for future clinical treatment.
{"title":"Modified Ce/Zr-MOF Nanoparticles Loaded with Curcumin for Alzheimer's Disease via Multifunctional Modulation.","authors":"Yan Yang, Yiling Wang, Xinran Jiang, Jiahao Mi, Dizhang Ge, Yuna Tong, Yuxuan Zhu","doi":"10.2147/IJN.S479242","DOIUrl":"10.2147/IJN.S479242","url":null,"abstract":"<p><strong>Introduction: </strong>Alzheimer's disease (AD), a neurodegenerative condition, stands as the most prevalent form of dementia. Its complex pathological mechanisms and the formidable blood-brain barrier (BBB) pose significant challenges to current treatment approaches. Oxidative stress is recognized as a central factor in AD, underscoring the importance of antioxidative strategies in its treatment. In this study, we developed a novel brain-targeted nanoparticle, Ce/Zr-MOF@Cur-Lf, for AD therapy.</p><p><strong>Methods: </strong>Layer-by-layer self-assembly technology was used to prepare Ce/Zr-MOF@Cur-Lf. In addition, the effect on the intracellular reactive oxygen species level, the uptake effect by PC12 and bEnd.3 cells and the in vitro BBB permeation effect were investigated. Finally, the mouse AD model was established by intrahippocampal injection of Aβ<sub>1-42</sub>, and the in vivo biodistribution, AD therapeutic effect and biosafety of the nanoparticles were researched at the animal level.</p><p><strong>Results: </strong>As anticipated, Ce/Zr-MOF@Cur-Lf demonstrated efficient BBB penetration and uptake by PC12 cells, leading to attenuation of H<sub>2</sub>O<sub>2</sub>-induced oxidative damage. Moreover, intravenous administration of Ce/Zr-MOF@Cur-Lf resulted in rapid brain access and improvement of various pathological features of AD, including neuronal damage, amyloid-β deposition, dysregulated central cholinergic system, oxidative stress, and neuroinflammation.</p><p><strong>Conclusion: </strong>Overall, Ce/Zr-MOF@Cur-Lf represents a promising approach for precise brain targeting and multi-target mechanisms in AD therapy, potentially serving as a viable option for future clinical treatment.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11444058/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142361477","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 : 2024-09-25eCollection Date: 2024-01-01DOI: 10.2147/IJN.S482329
Lin-Zhu Zhang, Rui-Jie Du, Duo Wang, Juan Qin, Chao Yu, Lei Zhang, Hai-Dong Zhu
With the in-depth knowledge of the pathological and physiological characteristics of the intestinal barrier-portal vein/intestinal lymphatic vessels-systemic circulation axis, oral targeted drug delivery is frequently being renewed. With many advantages, such as high safety, convenient administration, and good patient compliance, many researchers have begun to explore targeted drug delivery from intravenous injections to oral administration. Over the past few decades, the fields of materials science and nanomedicine have produced various drug delivery platforms that hold great potential in overcoming the multiple barriers associated with oral drug delivery. However, the oral transport of particles into the systemic circulation is extremely difficult due to immune rejection and biochemical invasion in the intestine, which limits absorption and entry into the bloodstream. The feasibility of the oral delivery of targeted drugs to sites outside the gastrointestinal tract (GIT) is unknown. This article reviews the biological barriers to drug absorption, the in vivo fate and transport mechanisms of drug carriers, the theoretical basis for oral administration, and the impact of carrier structural evolution on oral administration to achieve this goal. Finally, this article reviews the characteristics of different nano-delivery systems that can enhance the bioavailability of oral therapeutics and highlights their applications in the efficient creation of oral anticancer nanomedicines.
{"title":"Enteral Route Nanomedicine for Cancer Therapy.","authors":"Lin-Zhu Zhang, Rui-Jie Du, Duo Wang, Juan Qin, Chao Yu, Lei Zhang, Hai-Dong Zhu","doi":"10.2147/IJN.S482329","DOIUrl":"10.2147/IJN.S482329","url":null,"abstract":"<p><p>With the in-depth knowledge of the pathological and physiological characteristics of the intestinal barrier-portal vein/intestinal lymphatic vessels-systemic circulation axis, oral targeted drug delivery is frequently being renewed. With many advantages, such as high safety, convenient administration, and good patient compliance, many researchers have begun to explore targeted drug delivery from intravenous injections to oral administration. Over the past few decades, the fields of materials science and nanomedicine have produced various drug delivery platforms that hold great potential in overcoming the multiple barriers associated with oral drug delivery. However, the oral transport of particles into the systemic circulation is extremely difficult due to immune rejection and biochemical invasion in the intestine, which limits absorption and entry into the bloodstream. The feasibility of the oral delivery of targeted drugs to sites outside the gastrointestinal tract (GIT) is unknown. This article reviews the biological barriers to drug absorption, the in vivo fate and transport mechanisms of drug carriers, the theoretical basis for oral administration, and the impact of carrier structural evolution on oral administration to achieve this goal. Finally, this article reviews the characteristics of different nano-delivery systems that can enhance the bioavailability of oral therapeutics and highlights their applications in the efficient creation of oral anticancer nanomedicines.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11439897/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346217","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 : 2024-09-25eCollection Date: 2024-01-01DOI: 10.2147/IJN.S477835
Yan Yan, Hanqing Cai, Maoguang Yang
The endocrine system regulates many biological systems, and disruptions may result in disorders, such as diabetes, thyroid dysfunction, Cushing's syndrome, and obesity. The total incidence of endocrine illnesses was found to be 47.4%, excluding type 2 diabetes mellitus, with a significant frequency of newly diagnosed endocrine disorders. Nanotechnology manipulates particles at the atomic and molecular levels, opening up new paths for studying disease etiology and therapeutic alternatives. The goal of using nanomaterials in the treatment of endocrine illnesses is to create endogenous nano-biosensors that can detect even modest changes in hormone levels and react spontaneously to restore normal function. The size and surface characteristics of nanoparticles enhances the sensitivity in nano-sensors and are functionalized for targeted drug delivery. Nano-sized carriers composed of lipids, polymers, carbon, or metals have been shown to work much better than standard drug delivery methods. Nanoparticles (NPs) offer various advantages over current methods for diagnosing and treating endocrine disorders, acting as hydrogels for insulin delivery and wound healing. Incorporating selenium NPs into inorganic nanoparticles enhances their bioactivity and targeted delivery. Gold NPs show a promising precise insulin delivery. Mesoporous silica NPs maintain glycemic level effectively and lipid and polymeric NPs protect drugs from degradation in the gastrointestinal tract. Carbon nanotubes (CNTs) have become popular in thyroid surgeries. These characteristics make nanoparticles valuable for developing effective diagnostic and therapeutic systems. NP-based medicines have been thoroughly researched in order to identify the beginning point for the creation of theranostics, which may function in two ways: as imaging agents or therapeutics. The study posits that nanotechnology bridges diagnostics and therapies, potentially revolutionizing endocrine disorder treatments. This review delves into nanotechnology techniques, emphasizing their applications in diagnosing and treating diabetes mellitus.
{"title":"The Application of Nanotechnology for the Diagnosis and Treatment of Endocrine Disorders: A Review of Current Trends, Toxicology and Future Perspective.","authors":"Yan Yan, Hanqing Cai, Maoguang Yang","doi":"10.2147/IJN.S477835","DOIUrl":"https://doi.org/10.2147/IJN.S477835","url":null,"abstract":"<p><p>The endocrine system regulates many biological systems, and disruptions may result in disorders, such as diabetes, thyroid dysfunction, Cushing's syndrome, and obesity. The total incidence of endocrine illnesses was found to be 47.4%, excluding type 2 diabetes mellitus, with a significant frequency of newly diagnosed endocrine disorders. Nanotechnology manipulates particles at the atomic and molecular levels, opening up new paths for studying disease etiology and therapeutic alternatives. The goal of using nanomaterials in the treatment of endocrine illnesses is to create endogenous nano-biosensors that can detect even modest changes in hormone levels and react spontaneously to restore normal function. The size and surface characteristics of nanoparticles enhances the sensitivity in nano-sensors and are functionalized for targeted drug delivery. Nano-sized carriers composed of lipids, polymers, carbon, or metals have been shown to work much better than standard drug delivery methods. Nanoparticles (NPs) offer various advantages over current methods for diagnosing and treating endocrine disorders, acting as hydrogels for insulin delivery and wound healing. Incorporating selenium NPs into inorganic nanoparticles enhances their bioactivity and targeted delivery. Gold NPs show a promising precise insulin delivery. Mesoporous silica NPs maintain glycemic level effectively and lipid and polymeric NPs protect drugs from degradation in the gastrointestinal tract. Carbon nanotubes (CNTs) have become popular in thyroid surgeries. These characteristics make nanoparticles valuable for developing effective diagnostic and therapeutic systems. NP-based medicines have been thoroughly researched in order to identify the beginning point for the creation of theranostics, which may function in two ways: as imaging agents or therapeutics. The study posits that nanotechnology bridges diagnostics and therapies, potentially revolutionizing endocrine disorder treatments. This review delves into nanotechnology techniques, emphasizing their applications in diagnosing and treating diabetes mellitus.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11439355/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346223","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 : 2024-09-24eCollection Date: 2024-01-01DOI: 10.2147/IJN.S480901
Xiangan Gong, Qian Zhao, Huimin Zhang, Rui Liu, Jie Wu, Nanxin Zhang, Yuanxian Zou, Wen Zhao, Ran Huo, Rongtao Cui
Pathological scarring results from aberrant cutaneous wound healing due to the overactivation of biological behaviors of human skin fibroblasts, characterized by local inordinate inflammation, excessive extracellular matrix and collagen deposition. Yet, its underlying pathogenesis opinions vary, which could be caused by increased local mechanical tension, enhanced and continuous inflammation, gene mutation, as well as cellular metabolic disorder, etc. Metabolic reprogramming is the process by which the metabolic pattern of cells undergoes a systematic adjustment and transformation to adapt to the changes of the external environment and meet the needs of their growth and differentiation. Therefore, the abnormality of metabolic reprogramming in cells within wounds and scars attaches great importance to scar formation. Mesenchymal stem cells-derived exosomes (MSC-Exo) are the extracellular vesicles that play an important role in tissue repair, cancer treatment as well as immune and metabolic regulation. However, there is not a systematic work to detail the relevant studies. Herein, we gave a comprehensive summary of the existing research on three main metabolisms, including glycometabolism, lipid metabolism and amino acid metabolism, and MSC-Exo regulating metabolic reprogramming in wound healing and scar formation for further research reference.
{"title":"The Effects of Mesenchymal Stem Cells-Derived Exosomes on Metabolic Reprogramming in Scar Formation and Wound Healing.","authors":"Xiangan Gong, Qian Zhao, Huimin Zhang, Rui Liu, Jie Wu, Nanxin Zhang, Yuanxian Zou, Wen Zhao, Ran Huo, Rongtao Cui","doi":"10.2147/IJN.S480901","DOIUrl":"https://doi.org/10.2147/IJN.S480901","url":null,"abstract":"<p><p>Pathological scarring results from aberrant cutaneous wound healing due to the overactivation of biological behaviors of human skin fibroblasts, characterized by local inordinate inflammation, excessive extracellular matrix and collagen deposition. Yet, its underlying pathogenesis opinions vary, which could be caused by increased local mechanical tension, enhanced and continuous inflammation, gene mutation, as well as cellular metabolic disorder, etc. Metabolic reprogramming is the process by which the metabolic pattern of cells undergoes a systematic adjustment and transformation to adapt to the changes of the external environment and meet the needs of their growth and differentiation. Therefore, the abnormality of metabolic reprogramming in cells within wounds and scars attaches great importance to scar formation. Mesenchymal stem cells-derived exosomes (MSC-Exo) are the extracellular vesicles that play an important role in tissue repair, cancer treatment as well as immune and metabolic regulation. However, there is not a systematic work to detail the relevant studies. Herein, we gave a comprehensive summary of the existing research on three main metabolisms, including glycometabolism, lipid metabolism and amino acid metabolism, and MSC-Exo regulating metabolic reprogramming in wound healing and scar formation for further research reference.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438468/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346235","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}
Purpose: Renal cell carcinoma (RCC) is the most common and lethal type of urogenital cancer, with one-third of new cases presenting as metastatic RCC (mRCC), which, being the seventh most common cancer in men and the ninth in women, poses a significant challenge. For patients with poor prognosis, temsirolimus (TEM) has been approved for first-line therapy, possessing pharmacodynamic activities that block cancer cell growth and inhibit proliferation-associated proteins. However, TEM suffers from poor water solubility, low bioavailability, and systemic side effects. This study aims to develop a novel drug formulation for the treatment of RCC.
Methods: In this study, amphiphilic block copolymer (poly(ethylene glycol) monomethyl ether-poly(beta-amino ester)) (mPEG-PBAE) was utilized as a drug delivery vehicle and TEM-loaded micelles were prepared by thin-film hydration method by loading TEM inside the nanoparticles. Then, the molecular weight of mPEG-PBAE was controlled to make it realize hydrophobic-hydrophilic transition in the corresponding pH range thereby constructing pH-responsive TEM-loaded micelles. Characterization of pH-responsive TEM-loaded nanomicelles particle size, potential and micromorphology while its determination of drug-loading properties, in vitro release properties. Finally, pharmacodynamics and hepatorenal toxicity were further evaluated.
Results: TEM loading in mPEG-PBAE increased the solubility of TEM in water from 2.6 μg/mL to more than 5 mg/mL. The pH-responsive TEM-loaded nanomicelles were in the form of spheres or spheroidal shapes with an average particle size of 43.83 nm and a Zeta potential of 1.79 mV. The entrapment efficiency (EE) of pH-responsive TEM nanomicelles with 12.5% drug loading reached 95.27%. Under the environment of pH 6.7, the TEM was released rapidly within 12 h, and the release rate could reach 73.12% with significant pH-dependent characteristics. In vitro experiments showed that mPEG-PBAE preparation of TEM-loaded micelles had non-hemolytic properties and had significant inhibitory effects on cancer cells. In vivo experiments demonstrated that pH-responsive TEM-loaded micelles had excellent antitumor effects with significantly reduced liver and kidney toxicity.
Conclusion: In conclusion, we successfully prepared pH-responsive TEM-loaded micelles. The results showed that pH-responsive TEM-loaded micelles can achieve passive tumor targeting of TEM, and take advantage of the acidic conditions in tumor tissues to achieve rapid drug release.
目的:肾细胞癌(RCC)是泌尿生殖系统癌症中最常见、最致命的类型,三分之一的新病例表现为转移性肾细胞癌(mRCC)。对于预后不良的患者,替米考星(Temsirolimus,TEM)已被批准用于一线治疗,它具有阻断癌细胞生长和抑制增殖相关蛋白的药效学活性。然而,TEM 存在水溶性差、生物利用度低和全身副作用等问题。本研究旨在开发一种治疗 RCC 的新型药物制剂:方法:本研究采用两亲嵌段共聚物(聚乙二醇单甲醚-聚(β-氨基酯))(mPEG-PBAE)作为给药载体,通过薄膜水合法在纳米颗粒内负载 TEM,制备出负载 TEM 的胶束。然后,控制 mPEG-PBAE 的分子量,使其在相应的 pH 值范围内实现疏水-亲水转变,从而构建出 pH 响应型 TEM 负载胶束。对 pH 响应 TEM 负载纳米胶束的粒度、电位和微形貌进行表征,同时测定其药物负载特性和体外释放特性。最后,进一步评估了药效学和肝肾毒性:结果:在 mPEG-PBAE 中负载 TEM 可将 TEM 在水中的溶解度从 2.6 μg/mL 提高到 5 mg/mL 以上。具有 pH 响应的 TEM 负载纳米微球呈球形或球状,平均粒径为 43.83 nm,Zeta 电位为 1.79 mV。药物载量为 12.5% 的 pH 响应 TEM 纳米微球的包封效率(EE)达到 95.27%。在 pH 值为 6.7 的环境下,TEM 可在 12 小时内快速释放,释放率可达 73.12%,具有显著的 pH 依赖性。体外实验表明,mPEG-PBAE制备的TEM负载胶束具有非溶血性,对癌细胞有显著的抑制作用。体内实验表明,pH响应型TEM负载胶束具有良好的抗肿瘤效果,肝脏和肾脏毒性显著降低:总之,我们成功制备了 pH 响应 TEM 负载胶束。结果表明,pH响应型TEM载药胶束可实现TEM的被动肿瘤靶向,并利用肿瘤组织的酸性条件实现药物的快速释放。
{"title":"pH-Responsive Block Copolymer Micelles of Temsirolimus: Preparation, Characterization and Antitumor Activity Evaluation.","authors":"Ling Wang, Fangqing Cai, Yixuan Li, Xiaolan Lin, Yuting Wang, Weijie Liang, Caiyu Liu, Cunze Wang, Junshan Ruan","doi":"10.2147/IJN.S469913","DOIUrl":"https://doi.org/10.2147/IJN.S469913","url":null,"abstract":"<p><strong>Purpose: </strong>Renal cell carcinoma (RCC) is the most common and lethal type of urogenital cancer, with one-third of new cases presenting as metastatic RCC (mRCC), which, being the seventh most common cancer in men and the ninth in women, poses a significant challenge. For patients with poor prognosis, temsirolimus (TEM) has been approved for first-line therapy, possessing pharmacodynamic activities that block cancer cell growth and inhibit proliferation-associated proteins. However, TEM suffers from poor water solubility, low bioavailability, and systemic side effects. This study aims to develop a novel drug formulation for the treatment of RCC.</p><p><strong>Methods: </strong>In this study, amphiphilic block copolymer (poly(ethylene glycol) monomethyl ether-poly(beta-amino ester)) (mPEG-PBAE) was utilized as a drug delivery vehicle and TEM-loaded micelles were prepared by thin-film hydration method by loading TEM inside the nanoparticles. Then, the molecular weight of mPEG-PBAE was controlled to make it realize hydrophobic-hydrophilic transition in the corresponding pH range thereby constructing pH-responsive TEM-loaded micelles. Characterization of pH-responsive TEM-loaded nanomicelles particle size, potential and micromorphology while its determination of drug-loading properties, in vitro release properties. Finally, pharmacodynamics and hepatorenal toxicity were further evaluated.</p><p><strong>Results: </strong>TEM loading in mPEG-PBAE increased the solubility of TEM in water from 2.6 μg/mL to more than 5 mg/mL. The pH-responsive TEM-loaded nanomicelles were in the form of spheres or spheroidal shapes with an average particle size of 43.83 nm and a Zeta potential of 1.79 mV. The entrapment efficiency (EE) of pH-responsive TEM nanomicelles with 12.5% drug loading reached 95.27%. Under the environment of pH 6.7, the TEM was released rapidly within 12 h, and the release rate could reach 73.12% with significant pH-dependent characteristics. In vitro experiments showed that mPEG-PBAE preparation of TEM-loaded micelles had non-hemolytic properties and had significant inhibitory effects on cancer cells. In vivo experiments demonstrated that pH-responsive TEM-loaded micelles had excellent antitumor effects with significantly reduced liver and kidney toxicity.</p><p><strong>Conclusion: </strong>In conclusion, we successfully prepared pH-responsive TEM-loaded micelles. The results showed that pH-responsive TEM-loaded micelles can achieve passive tumor targeting of TEM, and take advantage of the acidic conditions in tumor tissues to achieve rapid drug release.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11430863/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346222","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 : 2024-09-23eCollection Date: 2024-01-01DOI: 10.2147/IJN.S476601
Wenjia Hou, Lan Shen, Yimin Zhu, Xuanjia Wang, Tianyu Du, Fang Yang, Yabin Zhu
Fullerenes hold tremendous potential as alternatives to conventional chemotherapy or radiotherapy for tumor treatment due to their abilities to photodynamically kill tumor cells, destroy the tumor vasculature, inhibit tumor metastasis and activate anti-tumor immune responses, while protecting normal tissue through antioxidative effects. The symmetrical hollow molecular structures of fullerenes with abundant C=C bonds allow versatile chemical modification with diverse functional groups, metal clusters and biomacromolecules to synthesize a wide range of fullerene derivatives with increased water solubility, improved biocompatibility, enhanced photodynamic properties and stronger targeting abilities. This review introduces the anti-tumor mechanisms of fullerenes and summarizes the most recent works on the functionalization of fullerenes and the application of fullerene derivatives in tumor treatment. This review aims to serve as a valuable reference for further development and clinical application of anti-tumor fullerene derivatives.
{"title":"Fullerene Derivatives for Tumor Treatment: Mechanisms and Application.","authors":"Wenjia Hou, Lan Shen, Yimin Zhu, Xuanjia Wang, Tianyu Du, Fang Yang, Yabin Zhu","doi":"10.2147/IJN.S476601","DOIUrl":"https://doi.org/10.2147/IJN.S476601","url":null,"abstract":"<p><p>Fullerenes hold tremendous potential as alternatives to conventional chemotherapy or radiotherapy for tumor treatment due to their abilities to photodynamically kill tumor cells, destroy the tumor vasculature, inhibit tumor metastasis and activate anti-tumor immune responses, while protecting normal tissue through antioxidative effects. The symmetrical hollow molecular structures of fullerenes with abundant C=C bonds allow versatile chemical modification with diverse functional groups, metal clusters and biomacromolecules to synthesize a wide range of fullerene derivatives with increased water solubility, improved biocompatibility, enhanced photodynamic properties and stronger targeting abilities. This review introduces the anti-tumor mechanisms of fullerenes and summarizes the most recent works on the functionalization of fullerenes and the application of fullerene derivatives in tumor treatment. This review aims to serve as a valuable reference for further development and clinical application of anti-tumor fullerene derivatives.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11430870/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346219","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}
Evodiamine (EVO) is a tryptamine indole alkaloid and the main active ingredient in Evodia rutaecarpa. In recent years, the antitumor, cardioprotective, anti-inflammatory, and anti-Alzheimer's disease effects of EVO have been reported. EVO exerts antitumor effects by inhibiting tumor cell activity and proliferation, blocking the cell cycle, promoting apoptosis and autophagy, and inhibiting the formation of the tumor microvasculature. However, EVO has poor solubility and low bioavailability. Several derivatives with high antitumor activity have been discovered through the structural optimization of EVO, and new drug delivery systems have been developed to improve the solubility and bioavailability of EVO. Current research found that EVO could have toxic effects, such as hepatotoxicity, nephrotoxicity, and cardiac toxicity. This article reviews the pharmacological activity, derivatives, drug delivery systems, toxicity, and pharmacokinetics of EVO and provides research ideas and references for its further in-depth development and clinical applications.
Evodiamine(EVO)是一种色胺吲哚生物碱,是Evodia rutaecarpa的主要活性成分。近年来,有关 EVO 的抗肿瘤、心脏保护、抗炎和抗老年痴呆症作用的报道屡见报端。EVO 通过抑制肿瘤细胞的活性和增殖、阻断细胞周期、促进细胞凋亡和自噬以及抑制肿瘤微血管的形成来发挥抗肿瘤作用。然而,EVO 溶解性差,生物利用度低。通过对 EVO 的结构优化,人们发现了几种具有高抗肿瘤活性的衍生物,并开发了新的给药系统来提高 EVO 的溶解性和生物利用度。目前的研究发现,EVO 可能具有毒性作用,如肝毒性、肾毒性和心脏毒性。本文综述了 EVO 的药理活性、衍生物、给药系统、毒性和药代动力学,并为其进一步深入开发和临床应用提供了研究思路和参考文献。
{"title":"Evodiamine: A Extremely Potential Drug Development Candidate of Alkaloids from <i>Evodia rutaecarpa</i>.","authors":"Longfei Lin, Yuling Liu, Ruying Tang, Shilan Ding, Hongmei Lin, Hui Li","doi":"10.2147/IJN.S459510","DOIUrl":"https://doi.org/10.2147/IJN.S459510","url":null,"abstract":"<p><p>Evodiamine (EVO) is a tryptamine indole alkaloid and the main active ingredient in <i>Evodia rutaecarpa</i>. In recent years, the antitumor, cardioprotective, anti-inflammatory, and anti-Alzheimer's disease effects of EVO have been reported. EVO exerts antitumor effects by inhibiting tumor cell activity and proliferation, blocking the cell cycle, promoting apoptosis and autophagy, and inhibiting the formation of the tumor microvasculature. However, EVO has poor solubility and low bioavailability. Several derivatives with high antitumor activity have been discovered through the structural optimization of EVO, and new drug delivery systems have been developed to improve the solubility and bioavailability of EVO. Current research found that EVO could have toxic effects, such as hepatotoxicity, nephrotoxicity, and cardiac toxicity. This article reviews the pharmacological activity, derivatives, drug delivery systems, toxicity, and pharmacokinetics of EVO and provides research ideas and references for its further in-depth development and clinical applications.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11430234/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346218","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 : 2024-09-23eCollection Date: 2024-01-01DOI: 10.2147/IJN.S472086
Mengyi Shen, Leyu Zhou, Xiaoli Fan, Ruiqi Wu, Shuyun Liu, Qiaoyu Deng, Yanyi Zheng, Jingping Liu, Li Yang
Background: Autoimmune hepatitis (AIH) is a serious liver disease characterized by immune disorders, particularly effector T-cell overactivation. This study aimed to explore the therapeutic effect and underlying mechanism of mesenchymal stem cell-derived extracellular vesicle (MSC-EV) treatment on CD4+ T-cell overactivation and liver injury in AIH.
Methods: The metabolic changes of CD4+ T cells were assayed in human AIH and mouse hepatitis models. The liver protective effect of MSC-EVs was evaluated by transaminase levels, liver histopathology and inflammation. The effect of MSC-EVs on the metabolic state of CD4+ T cells was also explored.
Results: Enhanced glycolysis (eg, ~1.5-fold increase in hexokinase 2 levels) was detected in the CD4+ T cells of AIH patient samples and mouse hepatitis models, whereas the inhibition of glycolysis decreased CD4+ T-cell activation (~1.8-fold decrease in CD69 levels) and AIH liver injury (~6-fold decrease in aminotransferase levels). MSC-EV treatment reduced CD4+ T-cell activation (~1.5-fold decrease in CD69 levels) and cytokine release (~5-fold decrease in IFN-γ levels) by reducing glycolysis (~3-fold decrease) while enhancing mitochondrial oxidative phosphorylation (~2-fold increase in maximal respiration) in such cells. The degree of liver damage in AIH mice was ameliorated after MSC-EV treatment (~5-fold decrease in aminotransferase levels). MSC-EVs carried abundant mitochondrial proteins and might transfer them to metabolically reprogram CD4+ T cells, whereas disrupting mitochondrial transfer impaired the therapeutic potency of MSC-EVs in activated CD4+ T cells.
Conclusion: Disordered glucose metabolism promotes CD4+ T-cell activation and associated inflammatory liver injury in AIH models, which can be reversed by MSC-EV therapy, and this effect is at least partially dependent on EV-mediated mitochondrial protein transfer between cells. This study highlights that MSC-EV therapy may represent a new avenue for treating autoimmune diseases such as AIH.
{"title":"Metabolic Reprogramming of CD4<sup>+</sup> T Cells by Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuates Autoimmune Hepatitis Through Mitochondrial Protein Transfer.","authors":"Mengyi Shen, Leyu Zhou, Xiaoli Fan, Ruiqi Wu, Shuyun Liu, Qiaoyu Deng, Yanyi Zheng, Jingping Liu, Li Yang","doi":"10.2147/IJN.S472086","DOIUrl":"https://doi.org/10.2147/IJN.S472086","url":null,"abstract":"<p><strong>Background: </strong>Autoimmune hepatitis (AIH) is a serious liver disease characterized by immune disorders, particularly effector T-cell overactivation. This study aimed to explore the therapeutic effect and underlying mechanism of mesenchymal stem cell-derived extracellular vesicle (MSC-EV) treatment on CD4<sup>+</sup> T-cell overactivation and liver injury in AIH.</p><p><strong>Methods: </strong>The metabolic changes of CD4<sup>+</sup> T cells were assayed in human AIH and mouse hepatitis models. The liver protective effect of MSC-EVs was evaluated by transaminase levels, liver histopathology and inflammation. The effect of MSC-EVs on the metabolic state of CD4<sup>+</sup> T cells was also explored.</p><p><strong>Results: </strong>Enhanced glycolysis (eg, ~1.5-fold increase in hexokinase 2 levels) was detected in the CD4<sup>+</sup> T cells of AIH patient samples and mouse hepatitis models, whereas the inhibition of glycolysis decreased CD4<sup>+</sup> T-cell activation (~1.8-fold decrease in CD69 levels) and AIH liver injury (~6-fold decrease in aminotransferase levels). MSC-EV treatment reduced CD4<sup>+</sup> T-cell activation (~1.5-fold decrease in CD69 levels) and cytokine release (~5-fold decrease in IFN-γ levels) by reducing glycolysis (~3-fold decrease) while enhancing mitochondrial oxidative phosphorylation (~2-fold increase in maximal respiration) in such cells. The degree of liver damage in AIH mice was ameliorated after MSC-EV treatment (~5-fold decrease in aminotransferase levels). MSC-EVs carried abundant mitochondrial proteins and might transfer them to metabolically reprogram CD4<sup>+</sup> T cells, whereas disrupting mitochondrial transfer impaired the therapeutic potency of MSC-EVs in activated CD4<sup>+</sup> T cells.</p><p><strong>Conclusion: </strong>Disordered glucose metabolism promotes CD4<sup>+</sup> T-cell activation and associated inflammatory liver injury in AIH models, which can be reversed by MSC-EV therapy, and this effect is at least partially dependent on EV-mediated mitochondrial protein transfer between cells. This study highlights that MSC-EV therapy may represent a new avenue for treating autoimmune diseases such as AIH.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11430536/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346221","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 : 2024-09-20eCollection Date: 2024-01-01DOI: 10.2147/IJN.S461167
Chloe Trayford, Dina M Ibrahim, Sabine van Rijt
Introduction: Oligonucleotide (ON) therapy is a promising treatment for a wide range of complex genetic disorders, but inefficient intracellular ON delivery has hindered clinical translation. Hollow silica nanoparticles (HSN) hold potential as effective ON delivery vehicles since ON can be encapsulated in the hollow core in situ where they are protected from degradation by eg nucleases. However, HSN must be modified to allow degradation and subsequent (sub)cellular ON release. In this report, we investigated the use of ion and fluorescent dye co-doping in the HSN silica matrix to enable HSN degradability and in vitro visualization.
Methods: HSN were core encapsulated with ON, doped with Ca2+, Cu2+, Zn2+, Se2+ and Sr2+ ions and co-condensed with rhodamine b isothiocyanate (RITC) by a modified reverse microemulsion method. HSN were physiochemically characterized and their biological activity such as uptake and toxicity were evaluated in mesenchymal stem cells (hMSCs).
Results: We successfully doped HSN with RITC and Ca2+, Cu2+, Zn2+ and Sr2+ ions. We observed that doping HSN with Ca2+ and Sr2+ enhanced RITC incorporation while ON encapsulation in HSN increased Cu2+ and Zn2+ doping efficiency. Moreover, our dual-doped HSN demonstrated controlled ON release in the presence of intracellular mimicking levels of glutathione (GSH) and limited release in the absence of GSH over 14 days. HSN were biocompatible in hMSCs up to 300 µg/mL except for Cu2+ doped HSNs which were cytotoxic even at ~10 µg/mL. HSN uptake was influenced by the dopant ion, DNA encapsulation, and HSN concentration, where Zn-HSN showed the lowest and Sr-HSN and Se-HSND, the highest uptake in hMSCs.
Conclusion: We report a straightforward one-pot procedure to create ion and fluorescent dye co-doped HSN that can efficiently incorporate ON, as promising new gene vectors.
{"title":"Ion Doped Hollow Silica Nanoparticles as Promising Oligonucleotide Delivery Systems to Mesenchymal Stem Cells.","authors":"Chloe Trayford, Dina M Ibrahim, Sabine van Rijt","doi":"10.2147/IJN.S461167","DOIUrl":"https://doi.org/10.2147/IJN.S461167","url":null,"abstract":"<p><strong>Introduction: </strong>Oligonucleotide (ON) therapy is a promising treatment for a wide range of complex genetic disorders, but inefficient intracellular ON delivery has hindered clinical translation. Hollow silica nanoparticles (HSN) hold potential as effective ON delivery vehicles since ON can be encapsulated in the hollow core in situ where they are protected from degradation by eg nucleases. However, HSN must be modified to allow degradation and subsequent (sub)cellular ON release. In this report, we investigated the use of ion and fluorescent dye co-doping in the HSN silica matrix to enable HSN degradability and in vitro visualization.</p><p><strong>Methods: </strong>HSN were core encapsulated with ON, doped with Ca<sup>2+</sup>, Cu<sup>2+</sup>, Zn<sup>2+</sup>, Se<sup>2+</sup> and Sr<sup>2+</sup> ions and co-condensed with rhodamine b isothiocyanate (RITC) by a modified reverse microemulsion method. HSN were physiochemically characterized and their biological activity such as uptake and toxicity were evaluated in mesenchymal stem cells (hMSCs).</p><p><strong>Results: </strong>We successfully doped HSN with RITC and Ca<sup>2+</sup>, Cu<sup>2+</sup>, Zn<sup>2+</sup> and Sr<sup>2+</sup> ions. We observed that doping HSN with Ca<sup>2+</sup> and Sr<sup>2+</sup> enhanced RITC incorporation while ON encapsulation in HSN increased Cu<sup>2+</sup> and Zn<sup>2+</sup> doping efficiency. Moreover, our dual-doped HSN demonstrated controlled ON release in the presence of intracellular mimicking levels of glutathione (GSH) and limited release in the absence of GSH over 14 days. HSN were biocompatible in hMSCs up to 300 µg/mL except for Cu<sup>2+</sup> doped HSNs which were cytotoxic even at ~10 µg/mL. HSN uptake was influenced by the dopant ion, DNA encapsulation, and HSN concentration, where Zn-HSN showed the lowest and Sr-HSN and Se-HSN<sub>D,</sub> the highest uptake in hMSCs.</p><p><strong>Conclusion: </strong>We report a straightforward one-pot procedure to create ion and fluorescent dye co-doped HSN that can efficiently incorporate ON, as promising new gene vectors.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11424689/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142346220","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}