Pub Date : 2024-11-10DOI: 10.1186/s12951-024-02956-w
Lizhi Wang, Junhao Xia, Xin Guan, Yang Song, Mengru Zhu, Fengya Wang, Baofeng Zhao, Lukuan Liu, Jing Liu
Extracellular vesicles (EVs) carry a variety of bioactive molecules and are becoming a promising alternative to cell therapy. Scale-up EV isolation is necessary for their functional studies and biological applications, while the traditional methods are challenged by low throughput, low yield, and potential damage. Herein, we developed an ion osmolarity-driven sequential concentration-enrichment strategy (IOSCE) for the EV isolation. IOSCE is composed of a novel superabsorbent polymers (SAPs) for EV concentration and a charged polymer for EV enrichment. Based on the driving force of ionic osmotic pressure, IOSCE can isolate EVs on a large scale from cell culture medium. The saturated water absorption capacity of IOSCE is 13.62 times higher than that of commercial SAPs. Compared with the ultracentrifugation method, IOSCE exhibited a 2.64 times higher yield (6.33 × 108 particles/mL). Moreover, the mesenchymal stem cell-derived EVs isolated using IOSCE demonstrate strong biological activity and can reduce neuroinflammation by affecting RNA metabolism and translation processes. IOSCE provides a cost-effective, high-throughput, and low-damage method for the scale up EV isolation, which is promising for disease diagnosis and treatment.
细胞外囊泡(EVs)携带多种生物活性分子,正在成为细胞疗法的一种有前途的替代方法。EVs的功能研究和生物应用离不开规模化的EVs分离,而传统方法面临着低通量、低产率和潜在损伤的挑战。在此,我们开发了一种离子渗透压驱动的连续浓缩富集策略(IOSCE),用于 EV 分离。IOSCE 由用于 EV 浓缩的新型超吸收聚合物(SAP)和用于 EV 富集的带电聚合物组成。基于离子渗透压的驱动力,IOSCE 可以从细胞培养基中大规模分离出 EV。IOSCE 的饱和吸水能力是商用 SAP 的 13.62 倍。与超速离心法相比,IOSCE 的产量高出 2.64 倍(6.33 × 108 颗粒/毫升)。此外,利用 IOSCE 分离的间充质干细胞衍生的 EVs 具有很强的生物活性,可通过影响 RNA 代谢和翻译过程减轻神经炎症。IOSCE提供了一种经济高效、高通量、低损伤的EV分离方法,有望用于疾病诊断和治疗。
{"title":"Ion osmolarity-driven sequential concentration-enrichment for the scale-up isolation of extracellular vesicles.","authors":"Lizhi Wang, Junhao Xia, Xin Guan, Yang Song, Mengru Zhu, Fengya Wang, Baofeng Zhao, Lukuan Liu, Jing Liu","doi":"10.1186/s12951-024-02956-w","DOIUrl":"10.1186/s12951-024-02956-w","url":null,"abstract":"<p><p>Extracellular vesicles (EVs) carry a variety of bioactive molecules and are becoming a promising alternative to cell therapy. Scale-up EV isolation is necessary for their functional studies and biological applications, while the traditional methods are challenged by low throughput, low yield, and potential damage. Herein, we developed an ion osmolarity-driven sequential concentration-enrichment strategy (IOSCE) for the EV isolation. IOSCE is composed of a novel superabsorbent polymers (SAPs) for EV concentration and a charged polymer for EV enrichment. Based on the driving force of ionic osmotic pressure, IOSCE can isolate EVs on a large scale from cell culture medium. The saturated water absorption capacity of IOSCE is 13.62 times higher than that of commercial SAPs. Compared with the ultracentrifugation method, IOSCE exhibited a 2.64 times higher yield (6.33 × 10<sup>8</sup> particles/mL). Moreover, the mesenchymal stem cell-derived EVs isolated using IOSCE demonstrate strong biological activity and can reduce neuroinflammation by affecting RNA metabolism and translation processes. IOSCE provides a cost-effective, high-throughput, and low-damage method for the scale up EV isolation, which is promising for disease diagnosis and treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"686"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550536/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-10DOI: 10.1186/s12951-024-02969-5
Chuanda Zhu, Jing Mu, Ling Liang
Protein drugs are of great importance in maintaining the normal functioning of living organisms. Indeed, they have been instrumental in combating tumors and genetic diseases for decades. Among these pharmaceutical agents, those that target intracellular components necessitate the use of therapeutic proteins to exert their effects within the targeted cells. However, the use of protein drugs is limited by their short half-life and potential adverse effects in the physiological environment. The advent of nanoparticles offers a promising avenue for prolonging the half-life of protein drugs. This is achieved by encapsulating proteins, thereby safeguarding their biological activity and ensuring precise delivery into cells. This nanomaterial-based intracellular protein drug delivery system mitigates the rapid hydrolysis and unwarranted diffusion of proteins, thereby minimizing potential side effects and circumventing the limitations inherent in traditional techniques like electroporation. This review examines established protein drug delivery systems, including those based on polymers, liposomes, and protein nanoparticles. We delve into the operational principles and transport mechanisms of nanocarriers, discussing the various considerations essential for designing cutting-edge delivery platforms. Additionally, we investigate innovative designs and applications of traditional cytosolic protein delivery systems in medical research and clinical practice, particularly in areas like tumor treatment, gene editing and fluorescence imaging. This review sheds light on the current restrictions of protein delivery systems and anticipates future research avenues, aiming to foster the continued advancement in this field.
{"title":"Nanocarriers for intracellular delivery of proteins in biomedical applications: strategies and recent advances.","authors":"Chuanda Zhu, Jing Mu, Ling Liang","doi":"10.1186/s12951-024-02969-5","DOIUrl":"10.1186/s12951-024-02969-5","url":null,"abstract":"<p><p>Protein drugs are of great importance in maintaining the normal functioning of living organisms. Indeed, they have been instrumental in combating tumors and genetic diseases for decades. Among these pharmaceutical agents, those that target intracellular components necessitate the use of therapeutic proteins to exert their effects within the targeted cells. However, the use of protein drugs is limited by their short half-life and potential adverse effects in the physiological environment. The advent of nanoparticles offers a promising avenue for prolonging the half-life of protein drugs. This is achieved by encapsulating proteins, thereby safeguarding their biological activity and ensuring precise delivery into cells. This nanomaterial-based intracellular protein drug delivery system mitigates the rapid hydrolysis and unwarranted diffusion of proteins, thereby minimizing potential side effects and circumventing the limitations inherent in traditional techniques like electroporation. This review examines established protein drug delivery systems, including those based on polymers, liposomes, and protein nanoparticles. We delve into the operational principles and transport mechanisms of nanocarriers, discussing the various considerations essential for designing cutting-edge delivery platforms. Additionally, we investigate innovative designs and applications of traditional cytosolic protein delivery systems in medical research and clinical practice, particularly in areas like tumor treatment, gene editing and fluorescence imaging. This review sheds light on the current restrictions of protein delivery systems and anticipates future research avenues, aiming to foster the continued advancement in this field.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"688"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552240/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-10DOI: 10.1186/s12951-024-02947-x
Mingping He, Ming Fang, Limin Fan, Alimujiang Maimaitijiang
{"title":"Correction: Preparation and characterization of BSA‑loaded liraglutide and platelet fragment nanoparticle delivery system for the treatment of diabetic atherosclerosis.","authors":"Mingping He, Ming Fang, Limin Fan, Alimujiang Maimaitijiang","doi":"10.1186/s12951-024-02947-x","DOIUrl":"https://doi.org/10.1186/s12951-024-02947-x","url":null,"abstract":"","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"693"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The characteristic neuropathology of Parkinson's disease (PD) involves the abnormal accumulation of phosphorylated α-synuclein (αSyn), as well as a significant decrease in neuromelanin (NM) levels within dopamine neurons (DaNs). Unlike αSyn aggregates, the relationship between NM levels and PD pathogenesis is not well understood. In this study, we engineered an E. coli MG1655 strain to produce exosomes containing melanin (E.melanin), and investigated its potential neuroprotective effects on DaNs in the context of PD. By employing a combination of cell cultures, biochemical studies, single nuclear RNA sequencing (snRNA seq), and various in vivo validations, we found that administration of E.melanin effectively alleviated DaNs loss and improved motor behavior impairments observed in both pharmacological and transgenic PD mouse models. Mechanistically, snRNA seq data suggested that E.melanin activated the PSAP-GPR37L1 signaling pathway specifically within astrocytes, leading to a reduction in astrocytic engulfment of synapses. Notably, activation of the GPR37L1 receptor using Tx14(A) peptide successfully rescued motor defects as well as protected against DaNs degeneration in mice with PD. Overall, our findings provide novel insights into understanding the molecular mechanisms underlying melanin's protective effects on DaNs in PD while offering potential strategies for manipulating and treating its pathophysiological progression.
帕金森病(PD)的特征性神经病理学包括磷酸化α-突触核蛋白(αSyn)的异常聚集,以及多巴胺神经元(DaNs)内神经髓鞘素(NM)水平的显著下降。与αSyn聚集不同,NM水平与帕金森病发病机制之间的关系尚不十分清楚。在本研究中,我们设计了一种大肠杆菌 MG1655 菌株来产生含有黑色素的外泌体(E.melanin),并研究了其在帕金森病中对 DaNs 的潜在神经保护作用。通过综合运用细胞培养、生化研究、单核糖核酸测序(snRNA seq)和各种体内验证,我们发现服用黑色素外泌体能有效缓解药物和转基因帕金森病小鼠模型中观察到的DaNs缺失,并改善运动行为障碍。从机理上讲,snRNA 序列数据表明,黑色素激活了星形胶质细胞内特异性的 PSAP-GPR37L1 信号通路,导致星形胶质细胞对突触的吞噬减少。值得注意的是,使用 Tx14(A) 肽激活 GPR37L1 受体成功地挽救了运动缺陷,并防止了帕金森病小鼠的 DaNs 退化。总之,我们的研究结果为了解黑色素对帕金森病 DaNs 的保护作用的分子机制提供了新的见解,同时也为操纵和治疗帕金森病的病理生理进展提供了潜在的策略。
{"title":"Genetically modified E. Coli secreting melanin (E.melanin) activates the astrocytic PSAP-GPR37L1 pathway and mitigates the pathogenesis of Parkinson's disease.","authors":"Weixian Kong, Yu Liu, Pu Ai, Yong Bi, Chaoguang Wei, Xiaoyang Guo, Zhenyu Cai, Ge Gao, Peng Hu, Jialin Zheng, Jianhui Liu, Minfeng Huo, Yuting Guan, Qihui Wu","doi":"10.1186/s12951-024-02955-x","DOIUrl":"10.1186/s12951-024-02955-x","url":null,"abstract":"<p><p>The characteristic neuropathology of Parkinson's disease (PD) involves the abnormal accumulation of phosphorylated α-synuclein (αSyn), as well as a significant decrease in neuromelanin (NM) levels within dopamine neurons (DaNs). Unlike αSyn aggregates, the relationship between NM levels and PD pathogenesis is not well understood. In this study, we engineered an E. coli MG1655 strain to produce exosomes containing melanin (E.melanin), and investigated its potential neuroprotective effects on DaNs in the context of PD. By employing a combination of cell cultures, biochemical studies, single nuclear RNA sequencing (snRNA seq), and various in vivo validations, we found that administration of E.melanin effectively alleviated DaNs loss and improved motor behavior impairments observed in both pharmacological and transgenic PD mouse models. Mechanistically, snRNA seq data suggested that E.melanin activated the PSAP-GPR37L1 signaling pathway specifically within astrocytes, leading to a reduction in astrocytic engulfment of synapses. Notably, activation of the GPR37L1 receptor using Tx14(A) peptide successfully rescued motor defects as well as protected against DaNs degeneration in mice with PD. Overall, our findings provide novel insights into understanding the molecular mechanisms underlying melanin's protective effects on DaNs in PD while offering potential strategies for manipulating and treating its pathophysiological progression.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"690"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552183/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-10DOI: 10.1186/s12951-024-02951-1
Ara Sargsian, Xanthippi Koutsoumpou, Hermon Girmatsion, Çan Egil, Kiana Buttiens, Carla Rios Luci, Stefaan J Soenen, Bella B Manshian
Cancer immunotherapy is often hindered by an immunosuppressive tumor microenvironment (TME). Various strategies are being evaluated to shift the TME from an immunologically 'cold' to 'hot' tumor and hereby improve current immune checkpoint blockades (ICB). One particular hot topic is the use of combination therapies. Here, we set out to screen a variety of metallic nanoparticles and explored their in vitro toxicity against a series of tumor and non-tumor cell lines. For silver nanoparticles, we also explored the effects of core size and surface chemistry on cytotoxicity. Ag-citrate-5 nm nanoparticles were found to induce high cytotoxicity in Renca cells through excessive generation of reactive oxygen species (ROS) and significantly increased cytokine production. The induced toxicity resulted in a shift of the immunogenic cell death (ICD) marker calreticulin to the cell surface in vitro and in vivo. Subcutaneous Renca tumors were treated with anti-PD1 or in combination with Ag-citrate-5 nm. The combination group resulted in significant reduction in tumor size, increased necrosis, and immune cell infiltration at the tumor site. Inhibition of cytotoxic CD8 + T cells confirmed the involvement of these cells in the observed therapeutic effects. Our results suggest that Ag-citrate-5 nm is able to promote immune cell influx and increase tumor responsiveness to ICB therapies.
{"title":"Silver nanoparticle induced immunogenic cell death can improve immunotherapy.","authors":"Ara Sargsian, Xanthippi Koutsoumpou, Hermon Girmatsion, Çan Egil, Kiana Buttiens, Carla Rios Luci, Stefaan J Soenen, Bella B Manshian","doi":"10.1186/s12951-024-02951-1","DOIUrl":"10.1186/s12951-024-02951-1","url":null,"abstract":"<p><p>Cancer immunotherapy is often hindered by an immunosuppressive tumor microenvironment (TME). Various strategies are being evaluated to shift the TME from an immunologically 'cold' to 'hot' tumor and hereby improve current immune checkpoint blockades (ICB). One particular hot topic is the use of combination therapies. Here, we set out to screen a variety of metallic nanoparticles and explored their in vitro toxicity against a series of tumor and non-tumor cell lines. For silver nanoparticles, we also explored the effects of core size and surface chemistry on cytotoxicity. Ag-citrate-5 nm nanoparticles were found to induce high cytotoxicity in Renca cells through excessive generation of reactive oxygen species (ROS) and significantly increased cytokine production. The induced toxicity resulted in a shift of the immunogenic cell death (ICD) marker calreticulin to the cell surface in vitro and in vivo. Subcutaneous Renca tumors were treated with anti-PD1 or in combination with Ag-citrate-5 nm. The combination group resulted in significant reduction in tumor size, increased necrosis, and immune cell infiltration at the tumor site. Inhibition of cytotoxic CD8 + T cells confirmed the involvement of these cells in the observed therapeutic effects. Our results suggest that Ag-citrate-5 nm is able to promote immune cell influx and increase tumor responsiveness to ICB therapies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"691"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552147/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142621680","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pancreatic ductal adenocarcinoma (PDAC), characterized by a dense extracellular matrix (ECM), presents significant therapeutic challenges due to its poor prognosis and high resistance to chemotherapy. Current chemodrugs and diagnostic agents largely fail to cross the barrier posed by the ECM, which severely limits the PDAC theranostics. This study introduces a novel theranostic strategy using thioether-hybridized hollow mesoporous organosilica nanoparticles (dsMNs) for the co-delivery of copper (Cu) and disulfiram (DSF), aiming to induce cuproptosis in PDAC cells. Our approach leverages the ECM-degrading enzyme collagenase, integrated with dsMNs, to enhance drug penetration by reducing matrix stiffness. Furthermore, the innovative use of a pancreatic cancer cell membrane coating on the nanoparticles enhances tumor targeting and stability (dsMCu-D@M-Co). The multifunctional platform not only facilitates deep drug penetration and triggers cuproptosis effectively but also utilizes the inherent properties of Cu to serve as a T1-weighted magnetic resonance imaging (MRI) contrast agent. In vitro and in vivo assessments demonstrate significant tumor size reduction in PDAC-bearing mice, highlighting the dual functionality of our platform in improving therapeutic efficacy and diagnostic precision. This integrated strategy represents a significant advancement in the management of PDAC, offering a promising new direction for overcoming one of the most lethal cancers.
{"title":"A collagenase-decorated Cu-based nanotheranostics: remodeling extracellular matrix for optimizing cuproptosis and MRI in pancreatic ductal adenocarcinoma.","authors":"Yining Wang, Qiaomei Zhou, Wangping Luo, Xiaoyan Yang, Jinguo Zhang, Yijie Lou, Jin Mao, Jiayi Chen, Fan Wu, Jue Hou, Guping Tang, Hongzhen Bai, Risheng Yu","doi":"10.1186/s12951-024-02968-6","DOIUrl":"10.1186/s12951-024-02968-6","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC), characterized by a dense extracellular matrix (ECM), presents significant therapeutic challenges due to its poor prognosis and high resistance to chemotherapy. Current chemodrugs and diagnostic agents largely fail to cross the barrier posed by the ECM, which severely limits the PDAC theranostics. This study introduces a novel theranostic strategy using thioether-hybridized hollow mesoporous organosilica nanoparticles (dsMNs) for the co-delivery of copper (Cu) and disulfiram (DSF), aiming to induce cuproptosis in PDAC cells. Our approach leverages the ECM-degrading enzyme collagenase, integrated with dsMNs, to enhance drug penetration by reducing matrix stiffness. Furthermore, the innovative use of a pancreatic cancer cell membrane coating on the nanoparticles enhances tumor targeting and stability (dsMCu-D@M-Co). The multifunctional platform not only facilitates deep drug penetration and triggers cuproptosis effectively but also utilizes the inherent properties of Cu to serve as a T1-weighted magnetic resonance imaging (MRI) contrast agent. In vitro and in vivo assessments demonstrate significant tumor size reduction in PDAC-bearing mice, highlighting the dual functionality of our platform in improving therapeutic efficacy and diagnostic precision. This integrated strategy represents a significant advancement in the management of PDAC, offering a promising new direction for overcoming one of the most lethal cancers.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"689"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552245/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-10DOI: 10.1186/s12951-024-02957-9
Jorge L Mejia-Mendez, Edwin E Reza-Zaldívar, A Sanchez-Martinez, O Ceballos-Sanchez, Diego E Navarro-López, L Marcelo Lozano, Juan Armendariz-Borunda, Naveen Tiwari, Daniel A Jacobo-Velázquez, Gildardo Sanchez-Ante, Edgar R López-Mena
Background: Lanthanide-based nanomaterials offer a promising alternative for cancer therapy because of their selectivity and effectiveness, which can be modified and predicted by leveraging the improved accuracy and enhanced decision-making of machine learning (ML) modeling.
Methods: In this study, erbium (Er3+) and ytterbium (Yb3+) were used to dope zinc oxide (ZnO) nanoparticles (NPs). Various characterization techniques and biological assays were employed to investigate the physicochemical and optical properties of the (Er, Yb)-doped ZnO NPs, revealing the influence of the lanthanide elements.
Results: The (Er, Yb)-doped ZnO NPs exhibited laminar-type morphologies, negative surface charges, and optical bandgaps that vary with the presence of Er3+ and Yb3+. The incorporation of lanthanide ions reduced the cytotoxicity activity of ZnO against HEPG-2, CACO-2, and U87 cell lines. Conversely, doping with Er3+ and Yb3+ enhanced the antioxidant activity of the ZnO against DPPH, ABTS, and H2O2 radicals. The extra tree (ET) and random forest (RF) models predicted the relevance of the characterization results vis-à-vis the cytotoxic properties of the synthesized NPs.
Conclusion: This study demonstrates, for the first time, the synthesis of ZnO NPs doped with Er and Yb via a solution polymerization route. According to characterization results, it was unveiled that the effect of optical bandgap variations influenced the cytotoxic performance of the developed lanthanide-doped ZnO NPs, being the undoped ZnO NPs the most cytotoxic ones. The presence alone or in combination of Er and Yb enhanced their scavenging capacity. ML models such as ET and RF efficiently demonstrated that the concentration and cell line type are key parameters that influence the cytotoxicity of (Er, Yb)-doped ZnO NPs achieving high accuracy rates of 98.96% and 98.67%, respectively. This study expands the knowledge of lanthanides as dopants of nanomaterials for biological and medical applications and supports their potential in cancer therapy by integrating robust ML approaches.
{"title":"Exploring the cytotoxic and antioxidant properties of lanthanide-doped ZnO nanoparticles: a study with machine learning interpretation.","authors":"Jorge L Mejia-Mendez, Edwin E Reza-Zaldívar, A Sanchez-Martinez, O Ceballos-Sanchez, Diego E Navarro-López, L Marcelo Lozano, Juan Armendariz-Borunda, Naveen Tiwari, Daniel A Jacobo-Velázquez, Gildardo Sanchez-Ante, Edgar R López-Mena","doi":"10.1186/s12951-024-02957-9","DOIUrl":"10.1186/s12951-024-02957-9","url":null,"abstract":"<p><strong>Background: </strong>Lanthanide-based nanomaterials offer a promising alternative for cancer therapy because of their selectivity and effectiveness, which can be modified and predicted by leveraging the improved accuracy and enhanced decision-making of machine learning (ML) modeling.</p><p><strong>Methods: </strong>In this study, erbium (Er<sup>3+</sup>) and ytterbium (Yb<sup>3+</sup>) were used to dope zinc oxide (ZnO) nanoparticles (NPs). Various characterization techniques and biological assays were employed to investigate the physicochemical and optical properties of the (Er, Yb)-doped ZnO NPs, revealing the influence of the lanthanide elements.</p><p><strong>Results: </strong>The (Er, Yb)-doped ZnO NPs exhibited laminar-type morphologies, negative surface charges, and optical bandgaps that vary with the presence of Er<sup>3+</sup> and Yb<sup>3+</sup>. The incorporation of lanthanide ions reduced the cytotoxicity activity of ZnO against HEPG-2, CACO-2, and U87 cell lines. Conversely, doping with Er<sup>3+</sup> and Yb<sup>3+</sup> enhanced the antioxidant activity of the ZnO against DPPH, ABTS, and H<sub>2</sub>O<sub>2</sub> radicals. The extra tree (ET) and random forest (RF) models predicted the relevance of the characterization results vis-à-vis the cytotoxic properties of the synthesized NPs.</p><p><strong>Conclusion: </strong>This study demonstrates, for the first time, the synthesis of ZnO NPs doped with Er and Yb via a solution polymerization route. According to characterization results, it was unveiled that the effect of optical bandgap variations influenced the cytotoxic performance of the developed lanthanide-doped ZnO NPs, being the undoped ZnO NPs the most cytotoxic ones. The presence alone or in combination of Er and Yb enhanced their scavenging capacity. ML models such as ET and RF efficiently demonstrated that the concentration and cell line type are key parameters that influence the cytotoxicity of (Er, Yb)-doped ZnO NPs achieving high accuracy rates of 98.96% and 98.67%, respectively. This study expands the knowledge of lanthanides as dopants of nanomaterials for biological and medical applications and supports their potential in cancer therapy by integrating robust ML approaches.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"687"},"PeriodicalIF":10.6,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552316/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-09DOI: 10.1186/s12951-024-02971-x
Lingtong Cheng, Jiemeng Tao, Peng Lu, Taibo Liang, Xutao Li, Dong Chang, Huan Su, Wei He, Zechao Qu, He Li, Wenjun Mu, Wei Zhang, Nan Liu, Jianfeng Zhang, Peijian Cao, Jingjing Jin
Background: Modulating the microbiome with nanomaterials has been proposed to improve plant growth, and reduce reliance on external inputs. Carbon Nanosol (CNS) was attracted for its potential to improve plant productivity. However, the mechanism between CNS and rhizosphere microorganisms remained largely elusive.
Results: Here, we tried to systematically explore the effects of CNS (600 and 1200 mg/L by concentration) on tobacco growth, soil physical properties, and root-associated microbiome. The influence of CNS on soil physicochemical properties and plant growth was significant and dose-dependent, leading to a 28.82% increase in biomass accumulation by 600 mg/L CNS. Comparison between the CNS-treated and control plants revealed significant differences in microbiome composition, including 1148 distinct ASVs (923 bacteria and 225 fungi), microbiome interactions, and metabolic function of root-associated microbiomes. Fungal and bacterial communities had different response patterns for CNS treatment, with phased and dose-dependent effects, with the most significant changes in microbial community structure observed at 1200 mg/L after 10 days of treatment. Microbial networks of CNS-treated plants had more nodes and edges, higher connectivity, and more hub microorganisms than those of control plants. Compared with control, CNS significantly elevated abundances of various bacterial biomarkers (such as Sphingomonas and Burkholderia) and fungi biomarkers (including Penicillium, Myceliophthora, and Talaromyces), which were potential plant-beneficial organisms. Functional prediction based on metagenomic data demonstrated pathways related to nutrient cycling being greatly enriched under CNS treatment. Furthermore, 391 culturable bacteria and 44 culturable fungi were isolated from soil and root samples. Among them, six bacteria and two fungi strains enriched upon CNS treatment were validated to have plant growth promotion effect, and two fungi (Cladosporium spp. and Talaromyces spp.) played their roles by mediating volatile organic compounds (VOCs). To some extent, the driving and shaping of the microbiome by CNS contributed to its impact on plant growth and development.
Conclusion: Our results revealed the key role of root-associated microbiota in mediating the interaction between CNS and plants, thus providing valuable insights and strategies for harnessing CNS to enhance plant growth.
{"title":"Manipulation in root-associated microbiome via carbon nanosol for plant growth improvements.","authors":"Lingtong Cheng, Jiemeng Tao, Peng Lu, Taibo Liang, Xutao Li, Dong Chang, Huan Su, Wei He, Zechao Qu, He Li, Wenjun Mu, Wei Zhang, Nan Liu, Jianfeng Zhang, Peijian Cao, Jingjing Jin","doi":"10.1186/s12951-024-02971-x","DOIUrl":"10.1186/s12951-024-02971-x","url":null,"abstract":"<p><strong>Background: </strong>Modulating the microbiome with nanomaterials has been proposed to improve plant growth, and reduce reliance on external inputs. Carbon Nanosol (CNS) was attracted for its potential to improve plant productivity. However, the mechanism between CNS and rhizosphere microorganisms remained largely elusive.</p><p><strong>Results: </strong>Here, we tried to systematically explore the effects of CNS (600 and 1200 mg/L by concentration) on tobacco growth, soil physical properties, and root-associated microbiome. The influence of CNS on soil physicochemical properties and plant growth was significant and dose-dependent, leading to a 28.82% increase in biomass accumulation by 600 mg/L CNS. Comparison between the CNS-treated and control plants revealed significant differences in microbiome composition, including 1148 distinct ASVs (923 bacteria and 225 fungi), microbiome interactions, and metabolic function of root-associated microbiomes. Fungal and bacterial communities had different response patterns for CNS treatment, with phased and dose-dependent effects, with the most significant changes in microbial community structure observed at 1200 mg/L after 10 days of treatment. Microbial networks of CNS-treated plants had more nodes and edges, higher connectivity, and more hub microorganisms than those of control plants. Compared with control, CNS significantly elevated abundances of various bacterial biomarkers (such as Sphingomonas and Burkholderia) and fungi biomarkers (including Penicillium, Myceliophthora, and Talaromyces), which were potential plant-beneficial organisms. Functional prediction based on metagenomic data demonstrated pathways related to nutrient cycling being greatly enriched under CNS treatment. Furthermore, 391 culturable bacteria and 44 culturable fungi were isolated from soil and root samples. Among them, six bacteria and two fungi strains enriched upon CNS treatment were validated to have plant growth promotion effect, and two fungi (Cladosporium spp. and Talaromyces spp.) played their roles by mediating volatile organic compounds (VOCs). To some extent, the driving and shaping of the microbiome by CNS contributed to its impact on plant growth and development.</p><p><strong>Conclusion: </strong>Our results revealed the key role of root-associated microbiota in mediating the interaction between CNS and plants, thus providing valuable insights and strategies for harnessing CNS to enhance plant growth.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"685"},"PeriodicalIF":10.6,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11549841/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622335","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-08DOI: 10.1186/s12951-024-02915-5
Lina Zhao, Xiangyu Deng, Yuqing Li, Qing Zhao, Lizhu Xiao, Jianjiang Xue, Anyi Chen, Wei Cheng, Min Zhao
CRISPR-Cas12a system, characterized by its precise sequence recognition and cleavage activity, has emerged as a powerful and programmable tool for molecular diagnostics. However, current CRISPR-Cas12a-based nucleic acid detection methods, particularly microRNA (miRNA) detection, necessitate additional bio-engineering strategies to exert control over Cas12a activity. Herein, we propose an engineered target-responsive hairpin DNA activator (TRHDA) to mediate forward tearing protospacer activated CRISPR-Cas12a system, which enables direct miRNA detection with high specificity and sensitivity. Target miRNA specifically binding to hairpin DNA can drive forward tearing protospacer in the stem sequence of hairpin structure, facilitating the complementarity between crRNA spacer and protospacer to activate Cas12a. Upon the hairpin DNA as input-responsive activator of Cas12a, a universal biosensing method enables the multiple miRNAs (miR-21, let-7a, miR-30a) detection and also has exceptional capability in identifying single-base mismatches and distinguishing homologous let-7/miR-30 family members. Besides, TRHDA-mediated Cas12a-powered biosensing has realized the evaluation of miR-21 expression levels in diverse cellular contexts by intracellular imaging. Considering the easy programmability of hairpin DNA in responsive region, this strategy could expand for the other target molecules detection (e.g., proteins, micromolecules, peptides, exosomes), which offers significant implications for biomarkers diagnostics utilizing the CRISPR-Cas12a system toolbox.
CRISPR-Cas12a 系统以其精确的序列识别和裂解活性为特点,已成为分子诊断领域一种强大的可编程工具。然而,目前基于CRISPR-Cas12a的核酸检测方法,尤其是microRNA(miRNA)检测,需要额外的生物工程策略来控制Cas12a的活性。在这里,我们提出了一种工程化的靶响应发夹DNA激活剂(TRHDA)来介导前向撕裂原位激活CRISPR-Cas12a系统,从而实现高特异性和高灵敏度的直接miRNA检测。目标 miRNA 与发夹 DNA 特异性结合后,可在发夹结构的茎序列中驱动前向撕裂原位聚合体,促进 crRNA spacer 与原位聚合体的互补,从而激活 Cas12a。利用发夹 DNA 作为 Cas12a 的输入响应激活剂,一种通用的生物传感方法可实现多种 miRNA(miR-21、let-7a、miR-30a)的检测,而且在识别单碱基错配和区分同源的 let-7/miR-30 家族成员方面也具有卓越的能力。此外,TRHDA 介导的 Cas12a 驱动生物传感技术通过细胞内成像实现了对不同细胞环境中 miR-21 表达水平的评估。考虑到发夹 DNA 在响应区的可编程性,这种策略可以扩展到其他目标分子(如蛋白质、微分子、肽、外泌体)的检测,这对利用 CRISPR-Cas12a 系统工具箱进行生物标记物诊断具有重要意义。
{"title":"Binding-driven forward tearing protospacer activated CRISPR-Cas12a system and applications for microRNA detection.","authors":"Lina Zhao, Xiangyu Deng, Yuqing Li, Qing Zhao, Lizhu Xiao, Jianjiang Xue, Anyi Chen, Wei Cheng, Min Zhao","doi":"10.1186/s12951-024-02915-5","DOIUrl":"10.1186/s12951-024-02915-5","url":null,"abstract":"<p><p>CRISPR-Cas12a system, characterized by its precise sequence recognition and cleavage activity, has emerged as a powerful and programmable tool for molecular diagnostics. However, current CRISPR-Cas12a-based nucleic acid detection methods, particularly microRNA (miRNA) detection, necessitate additional bio-engineering strategies to exert control over Cas12a activity. Herein, we propose an engineered target-responsive hairpin DNA activator (TRHDA) to mediate forward tearing protospacer activated CRISPR-Cas12a system, which enables direct miRNA detection with high specificity and sensitivity. Target miRNA specifically binding to hairpin DNA can drive forward tearing protospacer in the stem sequence of hairpin structure, facilitating the complementarity between crRNA spacer and protospacer to activate Cas12a. Upon the hairpin DNA as input-responsive activator of Cas12a, a universal biosensing method enables the multiple miRNAs (miR-21, let-7a, miR-30a) detection and also has exceptional capability in identifying single-base mismatches and distinguishing homologous let-7/miR-30 family members. Besides, TRHDA-mediated Cas12a-powered biosensing has realized the evaluation of miR-21 expression levels in diverse cellular contexts by intracellular imaging. Considering the easy programmability of hairpin DNA in responsive region, this strategy could expand for the other target molecules detection (e.g., proteins, micromolecules, peptides, exosomes), which offers significant implications for biomarkers diagnostics utilizing the CRISPR-Cas12a system toolbox.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"684"},"PeriodicalIF":10.6,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11545271/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-07DOI: 10.1186/s12951-024-02963-x
Lu Tan, Jiazhao Xie, Chenqi Liao, Xiaoguang Li, Weiyun Zhang, Changchun Cai, Liming Cheng, Xiong Wang
Background: Ferroptosis represents a nonapoptotic type of programmed cell death induced by excessive intracellular iron accumulation. Ferroptosis is an essential driver of the pathogenesis of Alzheimer's disease (AD). Tetrahedral framework nucleic acids (tFNAs) are a novel type of nanoparticle with superior antiapoptotic capacity and excellent biocompatibility. However, the effect of tFNAs on Aβ triggered ferroptosis, cognitive and synaptic impairments in AD remains unknown.
Methods: N2a cells were treated with Aβ combined with/without tFNAs. Cell viability and levels of Fe2+, lipid peroxidation, MDA, LDH, and GSH were examined. RNA sequencing was applied to explore dysregulated ferroptosis related genes. Seven-month-old APP/PS1 mice were intranasally administrated with tFNAs for two weeks. Fluorescence imaging was used to detect the tFNAs distribution in the brain. Novel object recognition (NOR) test followed by Morris water maze (MWM) was used to test the learning and memory performance of mice. Golgi staining, Western blot, and immunofluorescence staining were used to examine synaptic plasticity.
Results: tFNAs promoted cell viability and GSH levels, reduced the levels of Fe2+, lipid peroxidation, MDA, and LDH in N2a cells treated with Aβ. RNA sequencing revealed that tFNAs reversed the promotive effect of Aβ on ferroptosis driver Atf3 gene and suppressive effect on ferroptosis suppressors Rrm2 and Furin genes. Fluorescence imaging confirmed the brain infiltration of tFNAs. tFNAs rescued synaptic and memory impairments, and ferroptosis in seven-month-old APP/PS1 mice.
Conclusions: Collectively, tFNAs inhibited Aβ-mediated ferroptosis and ameliorated cognitive and synaptic impairments in AD mice. tFNAs may serve as novel option to deal with AD.
{"title":"Tetrahedral framework nucleic acids inhibit Aβ-mediated ferroptosis and ameliorate cognitive and synaptic impairments in Alzheimer's disease.","authors":"Lu Tan, Jiazhao Xie, Chenqi Liao, Xiaoguang Li, Weiyun Zhang, Changchun Cai, Liming Cheng, Xiong Wang","doi":"10.1186/s12951-024-02963-x","DOIUrl":"10.1186/s12951-024-02963-x","url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis represents a nonapoptotic type of programmed cell death induced by excessive intracellular iron accumulation. Ferroptosis is an essential driver of the pathogenesis of Alzheimer's disease (AD). Tetrahedral framework nucleic acids (tFNAs) are a novel type of nanoparticle with superior antiapoptotic capacity and excellent biocompatibility. However, the effect of tFNAs on Aβ triggered ferroptosis, cognitive and synaptic impairments in AD remains unknown.</p><p><strong>Methods: </strong>N2a cells were treated with Aβ combined with/without tFNAs. Cell viability and levels of Fe<sup>2+</sup>, lipid peroxidation, MDA, LDH, and GSH were examined. RNA sequencing was applied to explore dysregulated ferroptosis related genes. Seven-month-old APP/PS1 mice were intranasally administrated with tFNAs for two weeks. Fluorescence imaging was used to detect the tFNAs distribution in the brain. Novel object recognition (NOR) test followed by Morris water maze (MWM) was used to test the learning and memory performance of mice. Golgi staining, Western blot, and immunofluorescence staining were used to examine synaptic plasticity.</p><p><strong>Results: </strong>tFNAs promoted cell viability and GSH levels, reduced the levels of Fe<sup>2+</sup>, lipid peroxidation, MDA, and LDH in N2a cells treated with Aβ. RNA sequencing revealed that tFNAs reversed the promotive effect of Aβ on ferroptosis driver Atf3 gene and suppressive effect on ferroptosis suppressors Rrm2 and Furin genes. Fluorescence imaging confirmed the brain infiltration of tFNAs. tFNAs rescued synaptic and memory impairments, and ferroptosis in seven-month-old APP/PS1 mice.</p><p><strong>Conclusions: </strong>Collectively, tFNAs inhibited Aβ-mediated ferroptosis and ameliorated cognitive and synaptic impairments in AD mice. tFNAs may serve as novel option to deal with AD.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"22 1","pages":"682"},"PeriodicalIF":10.6,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11542373/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142591041","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}