Pub Date : 2024-10-28eCollection Date: 2024-01-01DOI: 10.34133/bmr.0096
Xuhan Liu, Qiuxia Min, Yihui Li, Siyuan Chen
Conventional aluminum adjuvants exhibit limited cellular immunity. Polyinosinic-polycytidylic acid (poly I:C) activates cytoplasmic retinoic acid-inducible gene-like receptor (RLR), triggering strong T cell activation and cellular responses. However, when applied as an adjuvant, its limited endocytosis and restricted cytoplasmic delivery diminish its effectiveness and increase its toxicity. Hybrid polymer-lipid nanoparticle (PLNP) possesses numerous benefits such as good stability, reduced drug leakage, simple fabrication, easy property modulation, and excellent reproducibility compared to the lipid nanoparticle or the polymeric vector. Here, we developed a novel cationic polymer-lipid hybrid adjuvant capable of incorporating poly I:C to enhance cellular immunity. The hepatitis B surface antigen (HBsAg) was immobilized onto poly I:C-incorprated PLNP (PPLNP) via electrostatic interactions, forming the HBsAg/PPLNP vaccine formulation. The PPLNP adjuvant largely enhanced the cellular endocytosis and cytoplasmic transport of poly I:C, activating RLR followed by promoting type I interferon (IFN) secretion. Meanwhile, PPLNP obviously enhanced the antigen uptake, prolonged antigen retention at the site of administration, and facilitated enhanced transport of antigens to lymph nodes. The HBsAg/PPLNP nanovaccine led to amplified concentrations of antigen-specific immunoglobulin G (IgG), IFN-γ, granzyme B, and an enhanced IgG2a/IgG1 ratio, alongside the FasL+/CD8+ T cell activation, favoring a T helper 1 (TH1)-driven immune response. PPLNP, distinguished by its biocompatibility, ease of fabrication, and effectiveness in augmenting cellular immunity, holds significant promise as a new adjuvant.
传统铝佐剂的细胞免疫功能有限。聚肌苷酸-聚胞苷酸(poly I:C)可激活细胞质视黄酸诱导基因样受体(RLR),引发强烈的 T 细胞激活和细胞反应。然而,在作为佐剂使用时,其有限的内吞作用和受限的细胞质递送会降低其有效性并增加其毒性。与脂质纳米粒子或聚合物载体相比,聚合物-脂质杂化纳米粒子(PLNP)具有稳定性好、减少药物渗漏、制造简单、性能易于调节、可重复性好等诸多优点。在这里,我们开发了一种新型阳离子聚合物-脂质混合佐剂,它能结合聚 I:C 增强细胞免疫力。乙型肝炎表面抗原(HBsAg)通过静电作用被固定在聚 I:C 嵌体聚合氯化萘(PPLNP)上,形成 HBsAg/PPLNP 疫苗制剂。PPLNP 佐剂在很大程度上增强了多聚 I:C 的细胞内吞和细胞质转运,激活了 RLR,继而促进了 I 型干扰素(IFN)的分泌。同时,PPLNP 明显增强了抗原的摄取,延长了抗原在给药部位的保留时间,并促进了抗原向淋巴结的转运。HBsAg/PPLNP 纳米疫苗使抗原特异性免疫球蛋白 G (IgG)、IFN-γ、颗粒酶 B 的浓度增加,IgG2a/IgG1 比率提高,同时 FasL+/CD8+ T 细胞活化,有利于 T 辅助细胞 1 (TH1) 驱动的免疫反应。PPLNP 具有生物相容性好、易于制造、可有效增强细胞免疫力等特点,有望成为一种新型佐剂。
{"title":"Enhanced Cellular Immunity for Hepatitis B Virus Vaccine: A Novel Polyinosinic-Polycytidylic Acid-Incorporated Adjuvant Leveraging Cytoplasmic Retinoic Acid-Inducible Gene-Like Receptor Activation and Increased Antigen Uptake.","authors":"Xuhan Liu, Qiuxia Min, Yihui Li, Siyuan Chen","doi":"10.34133/bmr.0096","DOIUrl":"10.34133/bmr.0096","url":null,"abstract":"<p><p>Conventional aluminum adjuvants exhibit limited cellular immunity. Polyinosinic-polycytidylic acid (poly I:C) activates cytoplasmic retinoic acid-inducible gene-like receptor (RLR), triggering strong T cell activation and cellular responses. However, when applied as an adjuvant, its limited endocytosis and restricted cytoplasmic delivery diminish its effectiveness and increase its toxicity. Hybrid polymer-lipid nanoparticle (PLNP) possesses numerous benefits such as good stability, reduced drug leakage, simple fabrication, easy property modulation, and excellent reproducibility compared to the lipid nanoparticle or the polymeric vector. Here, we developed a novel cationic polymer-lipid hybrid adjuvant capable of incorporating poly I:C to enhance cellular immunity. The hepatitis B surface antigen (HBsAg) was immobilized onto poly I:C-incorprated PLNP (PPLNP) via electrostatic interactions, forming the HBsAg/PPLNP vaccine formulation. The PPLNP adjuvant largely enhanced the cellular endocytosis and cytoplasmic transport of poly I:C, activating RLR followed by promoting type I interferon (IFN) secretion. Meanwhile, PPLNP obviously enhanced the antigen uptake, prolonged antigen retention at the site of administration, and facilitated enhanced transport of antigens to lymph nodes. The HBsAg/PPLNP nanovaccine led to amplified concentrations of antigen-specific immunoglobulin G (IgG), IFN-γ, granzyme B, and an enhanced IgG2a/IgG1 ratio, alongside the FasL<sup>+</sup>/CD8<sup>+</sup> T cell activation, favoring a T helper 1 (T<sub>H</sub>1)-driven immune response. PPLNP, distinguished by its biocompatibility, ease of fabrication, and effectiveness in augmenting cellular immunity, holds significant promise as a new adjuvant.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0096"},"PeriodicalIF":8.1,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11513446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142523835","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24eCollection Date: 2024-01-01DOI: 10.34133/bmr.0097
Raveena Nagareddy, Ja-Hae Kim, Ji-Hye Kim, Reju George Thomas, Kang-Ho Choi, Yong-Yeon Jeong
Cerebral ischemia impairs blood circulation, leading to elevated reactive oxygen species (ROS) production. A ROS-responsive delivery of drugs can enhance the therapeutic efficacy and minimize the side effects. There is insufficient evidence on the impact of ROS-responsive nanoparticles on ischemic stroke. We developed ROS-responsive chitosan-bilirubin (ChiBil) nanoparticles to target acute ischemic lesions and investigated the effect of atorvastatin-loaded ROS-responsive ChiBil. We randomly assigned rats with transient middle cerebral artery occlusion (MCAO) to 4 groups: saline, Statin, ChiBil, and ChiBil-Statin. These groups were treated daily via the tail vein for 7 d. Behavioral assessment, magnetic resonance (MR) imaging, evaluation of neuroinflammation, blood-brain barrier (BBB) integrity, apoptosis, and neurogenesis after stroke were conducted. In vitro, results showed nanoparticle uptake and reduced intracellular ROS, lipid peroxidation, and inflammatory cytokines (IL-6 and TNF-α). In vivo, results showed improved motor deficits and decreased infarct volumes on MR images in the ChiBil-Statin group compared with the Control group on day 7 (P < 0.05). Furthermore, the expression of inflammatory cytokines such as IL-1β and IL-6 was reduced in the ChiBil-Statin group compared with the Control group (P < 0.05). Improvements in BBB integrity, apoptosis, and neurogenesis were observed in the ChiBil-Statin group. The findings demonstrated that intravenous ROS-responsive multifunctional ChiBil-Statin could effectively deliver drugs to the ischemic brain, exerting marked synergistic pleiotropic neuroprotective effects. Therefore, ChiBil-Statin holds promise as a targeted therapy for ischemic vascular diseases characterized by increased ROS production, leading to new avenues for future research and potential clinical applications.
{"title":"Reactive Oxygen Species-Responsive Chitosan-Bilirubin Nanoparticles Loaded with Statin for Treatment of Cerebral Ischemia.","authors":"Raveena Nagareddy, Ja-Hae Kim, Ji-Hye Kim, Reju George Thomas, Kang-Ho Choi, Yong-Yeon Jeong","doi":"10.34133/bmr.0097","DOIUrl":"https://doi.org/10.34133/bmr.0097","url":null,"abstract":"<p><p>Cerebral ischemia impairs blood circulation, leading to elevated reactive oxygen species (ROS) production. A ROS-responsive delivery of drugs can enhance the therapeutic efficacy and minimize the side effects. There is insufficient evidence on the impact of ROS-responsive nanoparticles on ischemic stroke. We developed ROS-responsive chitosan-bilirubin (ChiBil) nanoparticles to target acute ischemic lesions and investigated the effect of atorvastatin-loaded ROS-responsive ChiBil. We randomly assigned rats with transient middle cerebral artery occlusion (MCAO) to 4 groups: saline, Statin, ChiBil, and ChiBil-Statin. These groups were treated daily via the tail vein for 7 d. Behavioral assessment, magnetic resonance (MR) imaging, evaluation of neuroinflammation, blood-brain barrier (BBB) integrity, apoptosis, and neurogenesis after stroke were conducted. In vitro, results showed nanoparticle uptake and reduced intracellular ROS, lipid peroxidation, and inflammatory cytokines (IL-6 and TNF-α). In vivo, results showed improved motor deficits and decreased infarct volumes on MR images in the ChiBil-Statin group compared with the Control group on day 7 (<i>P</i> < 0.05). Furthermore, the expression of inflammatory cytokines such as IL-1β and IL-6 was reduced in the ChiBil-Statin group compared with the Control group (<i>P</i> < 0.05). Improvements in BBB integrity, apoptosis, and neurogenesis were observed in the ChiBil-Statin group. The findings demonstrated that intravenous ROS-responsive multifunctional ChiBil-Statin could effectively deliver drugs to the ischemic brain, exerting marked synergistic pleiotropic neuroprotective effects. Therefore, ChiBil-Statin holds promise as a targeted therapy for ischemic vascular diseases characterized by increased ROS production, leading to new avenues for future research and potential clinical applications.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0097"},"PeriodicalIF":8.1,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11499631/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Klotho (KLO) is an anti-fibrotic protein expressed in the kidneys and has been decreasing in the development of renal fibrosis (RF). However, restoring the decline in KLO levels remains a great challenge during RF treatment. Herein, an injectable KLO-loaded chitosan (CS) hydrogel (KLO-Gel) is designed to achieve localized and prolonged release of KLO in the RF treatment. KLO-Gel was prepared by cross-linking CS with β-glycerophosphate (β-GP), followed by rapid (within 3 min) thermosensitive gelation at 37 °C. Furthermore, KLO-Gel exhibited a slow and sustained release (over 14 d) of KLO both in PBS and in the kidneys of mice with unilateral ureter obstruction (UUO). A single local injection of KLO-Gel into the renal capsule of UUO mice was more effective at reducing RF (i.e., maintaining renal function and tissue structure, alleviating extracellular matrix accumulation, and inhibiting the TGF-β1/Smad2/3 signaling pathway) over a 14-d period than daily intraperitoneal injections of free KLO or captopril. Crucially, CS was found to induce endogenous KLO secretion, highlighting the added value of using CS in RF treatment. Overall, this study demonstrated that KLO-Gel enhanced the anti-fibrotic efficacy of KLO while minimizing its off-target toxicity, and its clinical potential awaits further validation.
Klotho(KLO)是一种在肾脏中表达的抗纤维化蛋白,在肾纤维化(RF)的发展过程中一直在下降。然而,在肾纤维化治疗过程中,如何恢复 KLO 水平的下降仍然是一个巨大的挑战。本文设计了一种可注射的KLO负载壳聚糖(CS)水凝胶(KLO-Gel),以在射频治疗中实现KLO的局部和长效释放。KLO-Gel 的制备方法是将壳聚糖与 β-甘油磷酸酯(β-GP)交联,然后在 37 °C 温度下快速(3 分钟内)热敏凝胶化。此外,KLO-凝胶在 PBS 和单侧输尿管梗阻(UUO)小鼠的肾脏中都能缓慢、持续地释放 KLO(超过 14 天)。与每天腹腔注射游离 KLO 或卡托普利相比,14 天内向 UUO 小鼠肾囊局部注射一次 KLO-凝胶能更有效地减少 RF(即维持肾功能和组织结构、减轻细胞外基质堆积和抑制 TGF-β1/Smad2/3 信号通路)。最重要的是,研究发现 CS 能诱导内源性 KLO 分泌,这凸显了在射频治疗中使用 CS 的附加价值。总之,这项研究表明,KLO-凝胶增强了KLO的抗纤维化功效,同时将其脱靶毒性降至最低,其临床潜力有待进一步验证。
{"title":"An In Situ Sustained-Release Chitosan Hydrogel to Attenuate Renal Fibrosis by Retaining Klotho Expression.","authors":"Chenyang Li, Shuai Wang, Chenghui Liao, Ying Li, Yunfeng Zhou, Haiqiang Wu, Wei Xiong","doi":"10.34133/bmr.0099","DOIUrl":"https://doi.org/10.34133/bmr.0099","url":null,"abstract":"<p><p>Klotho (KLO) is an anti-fibrotic protein expressed in the kidneys and has been decreasing in the development of renal fibrosis (RF). However, restoring the decline in KLO levels remains a great challenge during RF treatment. Herein, an injectable KLO-loaded chitosan (CS) hydrogel (KLO-Gel) is designed to achieve localized and prolonged release of KLO in the RF treatment. KLO-Gel was prepared by cross-linking CS with β-glycerophosphate (β-GP), followed by rapid (within 3 min) thermosensitive gelation at 37 °C. Furthermore, KLO-Gel exhibited a slow and sustained release (over 14 d) of KLO both in PBS and in the kidneys of mice with unilateral ureter obstruction (UUO). A single local injection of KLO-Gel into the renal capsule of UUO mice was more effective at reducing RF (i.e., maintaining renal function and tissue structure, alleviating extracellular matrix accumulation, and inhibiting the TGF-β1/Smad2/3 signaling pathway) over a 14-d period than daily intraperitoneal injections of free KLO or captopril. Crucially, CS was found to induce endogenous KLO secretion, highlighting the added value of using CS in RF treatment. Overall, this study demonstrated that KLO-Gel enhanced the anti-fibrotic efficacy of KLO while minimizing its off-target toxicity, and its clinical potential awaits further validation.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0099"},"PeriodicalIF":8.1,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11499586/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Extensive phosphorylation at serine 22 (pSer22) on lamin A is the hallmark of cell mitosis, which contributes to the breakdown of nuclear envelope. In the interphase, pSer22 lamin A exists in low abundance and is involved in mechanotransduction, virus infection, and gene expression. Numerous evidences emerge to support lamin A regulation on cell function and fate by phosphorylation. However, live-cell imaging tools for visualizing the dynamics of pSer22 lamin A are yet to be established. Herein, we developed a novel lamin A phosphorylation sensor (LAPS) based on fluorescence resonance energy transfer (FRET) with high sensitivity and specificity. We observed the dynamic lamin A phosphorylation during the cell cycle progression in single living cells: the increase of pSer22 modification when cells entered the mitosis and recovered upon the mitosis exit. Our biosensor also showed the gradual reduction of pSer22 modification during cell adhesion and in response to hypotonic environment. By applying LAPS, we captured the propagation of pSer22 modification from inside to outside of the inner nuclear membrane, which further led to the breakdown of nuclear envelope. Meanwhile, we found the synchronous phosphorylation of pSer22 lamin A and H3S10ph at mitosis entry. Inhibition of Aurora B, the responsible kinase for H3S10ph, could shorten the mitotic period without obvious effect on the pSer22 modification level of lamin A. Thus, LAPS allows the spatiotemporal visualization of the lamin A pSer22, which will be useful for elucidating the molecular mechanisms underlying cell mitosis and mechanoresponsive processes.
片层 A 上丝氨酸 22(pSer22)处的广泛磷酸化是细胞有丝分裂的标志,它有助于核包膜的破裂。在细胞间期,pSer22 层片 A 的含量较低,它参与机械传导、病毒感染和基因表达。大量证据表明,层压板 A 通过磷酸化调控细胞功能和命运。然而,用于观察 pSer22 层片 A 动态的活细胞成像工具尚未建立。在此,我们开发了一种基于荧光共振能量转移(FRET)的新型层片A磷酸化传感器(LAPS),具有高灵敏度和特异性。我们在单个活细胞中观察到了细胞周期进展过程中的动态片层 A 磷酸化:当细胞进入有丝分裂期时,pSer22 修饰增加,而当有丝分裂期结束时,pSer22 修饰恢复。我们的生物传感器还显示,在细胞粘附和低渗环境中,pSer22 修饰逐渐减少。通过应用 LAPS,我们捕捉到了 pSer22 修饰从核内膜内部向外部的传播,这进一步导致了核膜的破裂。同时,我们还发现在有丝分裂开始时,pSer22 层析成像 A 和 H3S10ph 同步磷酸化。因此,LAPS可实现片层A pSer22的时空可视化,这将有助于阐明细胞有丝分裂和机械突触过程的分子机制。
{"title":"Genetically Encoded Fluorescence Resonance Energy Transfer Biosensor for Live-Cell Visualization of Lamin A Phosphorylation at Serine 22.","authors":"Jian Liu, Qianqian Li, Jinfeng Wang, Juhui Qiu, Jing Zhou, Qin Peng","doi":"10.34133/bmr.0091","DOIUrl":"https://doi.org/10.34133/bmr.0091","url":null,"abstract":"<p><p>Extensive phosphorylation at serine 22 (pSer22) on lamin A is the hallmark of cell mitosis, which contributes to the breakdown of nuclear envelope. In the interphase, pSer22 lamin A exists in low abundance and is involved in mechanotransduction, virus infection, and gene expression. Numerous evidences emerge to support lamin A regulation on cell function and fate by phosphorylation. However, live-cell imaging tools for visualizing the dynamics of pSer22 lamin A are yet to be established. Herein, we developed a novel lamin A phosphorylation sensor (LAPS) based on fluorescence resonance energy transfer (FRET) with high sensitivity and specificity. We observed the dynamic lamin A phosphorylation during the cell cycle progression in single living cells: the increase of pSer22 modification when cells entered the mitosis and recovered upon the mitosis exit. Our biosensor also showed the gradual reduction of pSer22 modification during cell adhesion and in response to hypotonic environment. By applying LAPS, we captured the propagation of pSer22 modification from inside to outside of the inner nuclear membrane, which further led to the breakdown of nuclear envelope. Meanwhile, we found the synchronous phosphorylation of pSer22 lamin A and H3S10ph at mitosis entry. Inhibition of Aurora B, the responsible kinase for H3S10ph, could shorten the mitotic period without obvious effect on the pSer22 modification level of lamin A. Thus, LAPS allows the spatiotemporal visualization of the lamin A pSer22, which will be useful for elucidating the molecular mechanisms underlying cell mitosis and mechanoresponsive processes.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0091"},"PeriodicalIF":8.1,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11494032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142516811","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.34133/bmr.0095
Eun Hye Kim, Youngri Ryu, Jiwoong Choi, Daeho Park, Jong Won Lee, Sung-Gil Chi, Sun Hwa Kim, Yoosoo Yang
Acquired resistance to chemotherapy is a major challenge in the treatment of triple-negative breast cancer (TNBC). Despite accumulated evidence showing microRNA-21 (miR-21) as a vital regulator of tumor progression, the role of miR-21 in modulating the multidrug resistance of TNBC remains obscure. In this study, we demonstrate that miR-21 affects chemoresistance in 4T1 TNBC cells in response to doxorubicin (DOX) by regulating the P-glycoprotein (P-gp) drug efflux pump. Overexpression of miR-21 in the 4T1 cells markedly reduced their sensitivity to DOX, impeding DOX-promoted cell death. We employed anti-miR-21 oligonucleotide conjugated with a PD-L1-binding peptide (P21) for targeted delivery to 4T1 tumor cells. The selective down-regulation of miR-21 in 4T1 TNBC led to the reversal of P-gp-mediated DOX resistance by up-regulating phosphatase and tensin homolog (PTEN). Our study highlights that miR-21 is a key regulator of drug efflux pumps in TNBC, and targeting miR-21 could enhance DOX sensitivity, offering a potential therapeutic option for patients with DOX-resistant TNBC.
{"title":"Targeting miR-21 to Overcome P-glycoprotein Drug Efflux in Doxorubicin-Resistant 4T1 Breast Cancer.","authors":"Eun Hye Kim, Youngri Ryu, Jiwoong Choi, Daeho Park, Jong Won Lee, Sung-Gil Chi, Sun Hwa Kim, Yoosoo Yang","doi":"10.34133/bmr.0095","DOIUrl":"10.34133/bmr.0095","url":null,"abstract":"<p><p>Acquired resistance to chemotherapy is a major challenge in the treatment of triple-negative breast cancer (TNBC). Despite accumulated evidence showing microRNA-21 (miR-21) as a vital regulator of tumor progression, the role of miR-21 in modulating the multidrug resistance of TNBC remains obscure. In this study, we demonstrate that miR-21 affects chemoresistance in 4T1 TNBC cells in response to doxorubicin (DOX) by regulating the P-glycoprotein (P-gp) drug efflux pump. Overexpression of miR-21 in the 4T1 cells markedly reduced their sensitivity to DOX, impeding DOX-promoted cell death. We employed anti-miR-21 oligonucleotide conjugated with a PD-L1-binding peptide (P21) for targeted delivery to 4T1 tumor cells. The selective down-regulation of miR-21 in 4T1 TNBC led to the reversal of P-gp-mediated DOX resistance by up-regulating phosphatase and tensin homolog (PTEN). Our study highlights that miR-21 is a key regulator of drug efflux pumps in TNBC, and targeting miR-21 could enhance DOX sensitivity, offering a potential therapeutic option for patients with DOX-resistant TNBC.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0095"},"PeriodicalIF":8.1,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11491560/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142482961","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The recent discovery of cuproptosis, a novel copper-ion-induced cell death pathway, has suggested the novel therapeutic potential for treating heterogeneous and drug-resistant cancers. Currently, copper ionophore-based therapeutics have been designed to treat cancers, utilizing copper ions as a strategic tool to impede tumor proliferation and promote cellular demise. However, limitations of copper ionophore-based therapies include nontargeted delivery of copper ions, low tumor accumulation, and short half-life. Strategies to enhance specificity involve targeting intracellular cuproptosis mechanisms using nanotechnology-based drugs. Additionally, the importance of exploring combination therapies cannot be overstated, as they are a key strategy in improving the efficacy of cancer treatments. Recent studies have reported the anticancer effects of nanomedicines that can induce cuproptosis of cancer both in vitro and in vivo. These cuproptosis-targeted nanomedicines could improve delivery efficiency with the pharmacokinetic properties of copper ion, resulting in increasing cuproptosis-based anticancer effects. This review will summarize the intricate nexus between copper ion and carcinogenesis, examining the pivotal roles of copper homeostasis and its dysregulation in cancer progression and fatality. Furthermore, we will introduce the latest advances in cuproptosis-targeted nanomedicines for cancer treatment. Finally, the challenges in cuproptosis-based nanomedicines will be discussed for future development directions.
{"title":"Copper-Based Nanomedicines for Cuproptosis-Mediated Effective Cancer Treatment.","authors":"Dahye Noh, Hokyung Lee, Sangmin Lee, In-Cheol Sun, Hong Yeol Yoon","doi":"10.34133/bmr.0094","DOIUrl":"10.34133/bmr.0094","url":null,"abstract":"<p><p>The recent discovery of cuproptosis, a novel copper-ion-induced cell death pathway, has suggested the novel therapeutic potential for treating heterogeneous and drug-resistant cancers. Currently, copper ionophore-based therapeutics have been designed to treat cancers, utilizing copper ions as a strategic tool to impede tumor proliferation and promote cellular demise. However, limitations of copper ionophore-based therapies include nontargeted delivery of copper ions, low tumor accumulation, and short half-life. Strategies to enhance specificity involve targeting intracellular cuproptosis mechanisms using nanotechnology-based drugs. Additionally, the importance of exploring combination therapies cannot be overstated, as they are a key strategy in improving the efficacy of cancer treatments. Recent studies have reported the anticancer effects of nanomedicines that can induce cuproptosis of cancer both in vitro and in vivo. These cuproptosis-targeted nanomedicines could improve delivery efficiency with the pharmacokinetic properties of copper ion, resulting in increasing cuproptosis-based anticancer effects. This review will summarize the intricate nexus between copper ion and carcinogenesis, examining the pivotal roles of copper homeostasis and its dysregulation in cancer progression and fatality. Furthermore, we will introduce the latest advances in cuproptosis-targeted nanomedicines for cancer treatment. Finally, the challenges in cuproptosis-based nanomedicines will be discussed for future development directions.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0094"},"PeriodicalIF":8.1,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11486892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142482959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-16eCollection Date: 2024-01-01DOI: 10.34133/bmr.0078
Arathy Vasukutty, Yeonwoo Jang, Dongwan Han, Hansoo Park, In-Kyu Park
The investigation into viral latency illuminates its pivotal role in the survival strategies of diverse viruses, including herpesviruses, HIV, and HPV. This underscores the delicate balance between dormancy and the potential for reactivation. The study explores the intricate mechanisms governing viral latency, encompassing episomal and proviral forms, and their integration with the host's genetic material. This integration provides resilience against cellular defenses, substantially impacting the host-pathogen dynamic, especially in the context of HIV, with implications for clinical outcomes. Addressing the challenge of eradicating latent reservoirs, this review underscores the potential of epigenetic and genetic interventions. It highlights the use of innovative nanocarriers like nanoparticles and liposomes for delivering latency-reversing agents. The precision in delivery, capacity to navigate biological barriers, and sustained drug release by these nanocarriers present a promising strategy to enhance therapeutic efficacy. The review further explores nanotechnology's integration in combating latent viral infections, leveraging nanoparticle-based platforms for drug delivery, gene editing, and vaccination. Advances in lipid-based nanocarriers, polymeric nanoparticles, and inorganic nanoparticles are discussed, illustrating their potential for targeted, efficient, and multifunctional antiviral therapy. By merging a deep understanding of viral latency's molecular underpinnings with nanotechnology's transformative capabilities, this review underscores the promise of novel therapeutic interventions. These interventions have great potential for managing persistent viral infections, heralding a new era in the fight against diseases such as neuroHIV/AIDS, herpes, and HPV.
{"title":"Navigating Latency-Inducing Viral Infections: Therapeutic Targeting and Nanoparticle Utilization.","authors":"Arathy Vasukutty, Yeonwoo Jang, Dongwan Han, Hansoo Park, In-Kyu Park","doi":"10.34133/bmr.0078","DOIUrl":"https://doi.org/10.34133/bmr.0078","url":null,"abstract":"<p><p>The investigation into viral latency illuminates its pivotal role in the survival strategies of diverse viruses, including herpesviruses, HIV, and HPV. This underscores the delicate balance between dormancy and the potential for reactivation. The study explores the intricate mechanisms governing viral latency, encompassing episomal and proviral forms, and their integration with the host's genetic material. This integration provides resilience against cellular defenses, substantially impacting the host-pathogen dynamic, especially in the context of HIV, with implications for clinical outcomes. Addressing the challenge of eradicating latent reservoirs, this review underscores the potential of epigenetic and genetic interventions. It highlights the use of innovative nanocarriers like nanoparticles and liposomes for delivering latency-reversing agents. The precision in delivery, capacity to navigate biological barriers, and sustained drug release by these nanocarriers present a promising strategy to enhance therapeutic efficacy. The review further explores nanotechnology's integration in combating latent viral infections, leveraging nanoparticle-based platforms for drug delivery, gene editing, and vaccination. Advances in lipid-based nanocarriers, polymeric nanoparticles, and inorganic nanoparticles are discussed, illustrating their potential for targeted, efficient, and multifunctional antiviral therapy. By merging a deep understanding of viral latency's molecular underpinnings with nanotechnology's transformative capabilities, this review underscores the promise of novel therapeutic interventions. These interventions have great potential for managing persistent viral infections, heralding a new era in the fight against diseases such as neuroHIV/AIDS, herpes, and HPV.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0078"},"PeriodicalIF":8.1,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11480834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142482960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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: 10.34133/bmr.0088
Hongye He, Feng Li, Rui Tang, Nianhong Wu, Ying Zhou, Yuting Cao, Can Wang, Li Wan, Yang Zhou, Hua Zhuang, Pan Li
[This corrects the article DOI: 10.34133/bmr.0064.].
[This corrects the article DOI: 10.34133/bmr.0064.].
{"title":"Erratum to \"Ultrasound Controllable Release of Proteolysis Targeting Chimeras for Triple-Negative Breast Cancer Treatment\".","authors":"Hongye He, Feng Li, Rui Tang, Nianhong Wu, Ying Zhou, Yuting Cao, Can Wang, Li Wan, Yang Zhou, Hua Zhuang, Pan Li","doi":"10.34133/bmr.0088","DOIUrl":"https://doi.org/10.34133/bmr.0088","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.34133/bmr.0064.].</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0088"},"PeriodicalIF":8.1,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11456695/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142395926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-03eCollection Date: 2024-01-01DOI: 10.34133/bmr.0081
Molly C McCloskey, S Danial Ahmad, Louis P Widom, Pelin Kasap, Benjamin D Gastfriend, Eric V Shusta, Sean P Palecek, Britta Engelhardt, Thomas R Gaborski, Jonathan Flax, Richard E Waugh, James L McGrath
Sepsis is the most lethal and expensive condition treated in intensive care units. Sepsis survivors frequently suffer long-term cognitive impairment, which has been linked to the breakdown of the blood-brain barrier (BBB) during a sepsis-associated "cytokine storm". Because animal models poorly recapitulate sepsis pathophysiology, human models are needed to understand sepsis-associated brain injury and to develop novel therapeutic strategies. With the concurrent emergence of tissue chip technologies and the maturation of protocols for human induced pluripotent stem cell (hiPSC), we can now develop advanced in vitro models of the human BBB and immune system to understand the relationship between systemic inflammation and brain injury. Here, we present a BBB model of the primary barrier developed on the μSiM (microphysiological system enabled by an ultrathin silicon nanomembrane) tissue chip platform. The model features isogenically matched hiPSC-derived extended endothelial culture method brain microvascular endothelial cell-like cells (EECM-BMEC-like cells) and brain pericyte-like cells (BPLCs) in a back-to-back coculture separated by the ultrathin (100 nm) membrane. Both endothelial monocultures and cocultures with pericytes responded to sepsis-like stimuli, with increased small-molecule permeability, although no differences were detected between culture conditions. Conversely, BPLC coculture reduced the number of neutrophils that crossed the EECM-BMEC-like cell monolayer under sepsis-like stimulation. Interestingly, this barrier protection was not seen when the stimulus originated from the tissue side. Our studies are consistent with the reported role for pericytes in regulating leukocyte trafficking during sepsis but indicate that EECM-BMEC-like cells alone are sufficient to maintain the restrictive small-molecule permeability of the BBB.
{"title":"Pericytes Enrich the Basement Membrane and Reduce Neutrophil Transmigration in an In Vitro Model of Peripheral Inflammation at the Blood-Brain Barrier.","authors":"Molly C McCloskey, S Danial Ahmad, Louis P Widom, Pelin Kasap, Benjamin D Gastfriend, Eric V Shusta, Sean P Palecek, Britta Engelhardt, Thomas R Gaborski, Jonathan Flax, Richard E Waugh, James L McGrath","doi":"10.34133/bmr.0081","DOIUrl":"10.34133/bmr.0081","url":null,"abstract":"<p><p>Sepsis is the most lethal and expensive condition treated in intensive care units. Sepsis survivors frequently suffer long-term cognitive impairment, which has been linked to the breakdown of the blood-brain barrier (BBB) during a sepsis-associated \"cytokine storm\". Because animal models poorly recapitulate sepsis pathophysiology, human models are needed to understand sepsis-associated brain injury and to develop novel therapeutic strategies. With the concurrent emergence of tissue chip technologies and the maturation of protocols for human induced pluripotent stem cell (hiPSC), we can now develop advanced in vitro models of the human BBB and immune system to understand the relationship between systemic inflammation and brain injury. Here, we present a BBB model of the primary barrier developed on the μSiM (microphysiological system enabled by an ultrathin silicon nanomembrane) tissue chip platform. The model features isogenically matched hiPSC-derived extended endothelial culture method brain microvascular endothelial cell-like cells (EECM-BMEC-like cells) and brain pericyte-like cells (BPLCs) in a back-to-back coculture separated by the ultrathin (100 nm) membrane. Both endothelial monocultures and cocultures with pericytes responded to sepsis-like stimuli, with increased small-molecule permeability, although no differences were detected between culture conditions. Conversely, BPLC coculture reduced the number of neutrophils that crossed the EECM-BMEC-like cell monolayer under sepsis-like stimulation. Interestingly, this barrier protection was not seen when the stimulus originated from the tissue side. Our studies are consistent with the reported role for pericytes in regulating leukocyte trafficking during sepsis but indicate that EECM-BMEC-like cells alone are sufficient to maintain the restrictive small-molecule permeability of the BBB.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0081"},"PeriodicalIF":8.1,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447289/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373756","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01eCollection Date: 2024-01-01DOI: 10.34133/bmr.0068
Sol Shin, Chan Ho Kim, Soyoung Son, Jae Ah Lee, Seunglee Kwon, Dong Gil You, Jungmi Lee, Jeongyun Kim, Dong-Gyu Jo, Hyewon Ko, Jae Hyung Park
The abnormal tumor vasculature acts as the physical and functional barrier to the infiltration and activity of effector T cells, leading to the low response rate of immune checkpoint inhibitors (ICIs). Herein, antiangiogenic extracellular vesicles that enable normalization of the tumor-associated vasculature were prepared to potentiate the efficacy of ICIs. Small extracellular vesicles were exploited as the delivery platform to protect the antiangiogenic protein, pigment epithelium-derived factor (PEDF), from proteolytic degradation. Along with the physicochemical characteristics of the PEDF-enriched extracellular vesicles (P-EVs), their inhibitory effects on migration, proliferation, and tube formation of endothelial cells were investigated in vitro. In tumor-bearing mice, it was confirmed that, compared to bare PEDFs, P-EVs efficiently reduced vessel leakiness, improved blood perfusion, and attenuated hypoxia. Consequently, when combined with anti-PD-1 antibodies, P-EVs remarkably augmented the antitumor immunity, as evidenced by increased infiltration of CD8+ T cells and reduced regulatory T cells. These results suggest that P-EVs are promising therapeutics for tumors refractory to ICIs.
{"title":"PEDF-Enriched Extracellular Vesicle for Vessel Normalization to Potentiate Immune Checkpoint Blockade Therapy.","authors":"Sol Shin, Chan Ho Kim, Soyoung Son, Jae Ah Lee, Seunglee Kwon, Dong Gil You, Jungmi Lee, Jeongyun Kim, Dong-Gyu Jo, Hyewon Ko, Jae Hyung Park","doi":"10.34133/bmr.0068","DOIUrl":"10.34133/bmr.0068","url":null,"abstract":"<p><p>The abnormal tumor vasculature acts as the physical and functional barrier to the infiltration and activity of effector T cells, leading to the low response rate of immune checkpoint inhibitors (ICIs). Herein, antiangiogenic extracellular vesicles that enable normalization of the tumor-associated vasculature were prepared to potentiate the efficacy of ICIs. Small extracellular vesicles were exploited as the delivery platform to protect the antiangiogenic protein, pigment epithelium-derived factor (PEDF), from proteolytic degradation. Along with the physicochemical characteristics of the PEDF-enriched extracellular vesicles (P-EVs), their inhibitory effects on migration, proliferation, and tube formation of endothelial cells were investigated in vitro. In tumor-bearing mice, it was confirmed that, compared to bare PEDFs, P-EVs efficiently reduced vessel leakiness, improved blood perfusion, and attenuated hypoxia. Consequently, when combined with anti-PD-1 antibodies, P-EVs remarkably augmented the antitumor immunity, as evidenced by increased infiltration of CD8<sup>+</sup> T cells and reduced regulatory T cells. These results suggest that P-EVs are promising therapeutics for tumors refractory to ICIs.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0068"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142362487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}