Pub Date : 2024-12-19eCollection Date: 2024-01-01DOI: 10.34133/bmr.0098
Junjia He, Luoqin Fu, Yeyu Shen, Yan Teng, Youming Huang, Xiaoxia Ding, Danfeng Xu, Hong Cui, Mingang Zhu, Jiahao Xie, Yue Su, Ting Li, Weitao Huang, Xiaozhou Mou, Qiong Bian, Yibin Fan
Ultraviolet (UV) irradiation leads to the degradation of the extracellular matrix and collagen, thereby accelerating skin aging and imposing substantial psychological burden on patients. Current anti-aging strategies are limited and often associated with high costs or strong side effects. Plant-derived extracellular vesicle-like nanovesicles, with advantages such as natural availability and cost-effectiveness, show potential in anti-aging interventions. This study extracted extracellular vesicle-like nanovesicle from Polygonum multiflorum (PMELNVs) and systematically investigated their composition and metabolic pathways, further examining their efficacy and underlying mechanisms in combating photoaging. Results revealed the excellent antioxidative properties of PMELNVs, alleviating UV-induced oxidative stress, inhibiting matrix metalloproteinase production, reducing extracellular matrix degradation, promoting collagen synthesis, and ultimately exerting anti-photoaging effects. Additionally, safety assessments demonstrated favorable biocompatibility of PMELNVs. This study provides novel evidence supporting PMELNVs' ability to resist photoaging by reducing oxidative stress and enhancing collagen expression, thereby offering potential as a new natural therapeutic agent against skin photoaging and promising a safer and more effective local anti-aging strategy.
{"title":"<i>Polygonum multiflorum</i> Extracellular Vesicle-Like Nanovesicle for Skin Photoaging Therapy.","authors":"Junjia He, Luoqin Fu, Yeyu Shen, Yan Teng, Youming Huang, Xiaoxia Ding, Danfeng Xu, Hong Cui, Mingang Zhu, Jiahao Xie, Yue Su, Ting Li, Weitao Huang, Xiaozhou Mou, Qiong Bian, Yibin Fan","doi":"10.34133/bmr.0098","DOIUrl":"10.34133/bmr.0098","url":null,"abstract":"<p><p>Ultraviolet (UV) irradiation leads to the degradation of the extracellular matrix and collagen, thereby accelerating skin aging and imposing substantial psychological burden on patients. Current anti-aging strategies are limited and often associated with high costs or strong side effects. Plant-derived extracellular vesicle-like nanovesicles, with advantages such as natural availability and cost-effectiveness, show potential in anti-aging interventions. This study extracted extracellular vesicle-like nanovesicle from <i>Polygonum multiflorum</i> (PMELNVs) and systematically investigated their composition and metabolic pathways, further examining their efficacy and underlying mechanisms in combating photoaging. Results revealed the excellent antioxidative properties of PMELNVs, alleviating UV-induced oxidative stress, inhibiting matrix metalloproteinase production, reducing extracellular matrix degradation, promoting collagen synthesis, and ultimately exerting anti-photoaging effects. Additionally, safety assessments demonstrated favorable biocompatibility of PMELNVs. This study provides novel evidence supporting PMELNVs' ability to resist photoaging by reducing oxidative stress and enhancing collagen expression, thereby offering potential as a new natural therapeutic agent against skin photoaging and promising a safer and more effective local anti-aging strategy.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0098"},"PeriodicalIF":8.1,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11658808/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142866582","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 : 2024-12-18eCollection Date: 2024-01-01DOI: 10.34133/bmr.0109
Jongdarm Yi, Yujin Byun, Seong Soo Kang, Kyung Mi Shim, Kwangsik Jang, Jae Young Lee
Background: Mesenchymal stem cells (MSCs) offer a promising avenue for cartilage regeneration; however, their therapeutic efficacy requires substantial improvement. Cell priming using electrical stimulation (ES) is a promising approach to augmenting the therapeutic potential of MSCs and has shown potential for various regenerative applications. This study aimed to promote the ES-mediated chondrogenic differentiation of human MSCs and facilitate the repair of injured articular cartilage. Methods: MSCs were subjected to ES under various conditions (e.g., voltage, frequency, and number of repetitions) to enhance their capability of chondrogenesis and cartilage regeneration. Chondrogenic differentiation of electrically primed MSCs (epMSCs) was assessed based on gene expression and sulfated glycosaminoglycan production, and epMSCs with hyaluronic acid were transplanted into a rat osteochondral defect model. Transcriptomic analysis was performed to determine changes in gene expression by ES. Results: epMSCs exhibited significantly increased chondrogenic gene expression and sulfated glycosaminoglycan production compared with those in unstimulated controls. Macroscopic and histological results showed that in vivo epMSC transplantation considerably enhanced cartilage regeneration. Furthermore, ES markedly altered the expression of numerous genes of MSCs, including those associated with the extracellular matrix, the Wnt signaling pathway, and cartilage development. Conclusion: ES can effectively prime MSCs to improve articular cartilage repair, offering a promising strategy for enhancing the efficacy of various MSC-based therapies.
{"title":"Enhanced Chondrogenic Differentiation of Electrically Primed Human Mesenchymal Stem Cells for the Regeneration of Osteochondral Defects.","authors":"Jongdarm Yi, Yujin Byun, Seong Soo Kang, Kyung Mi Shim, Kwangsik Jang, Jae Young Lee","doi":"10.34133/bmr.0109","DOIUrl":"10.34133/bmr.0109","url":null,"abstract":"<p><p><b>Background:</b> Mesenchymal stem cells (MSCs) offer a promising avenue for cartilage regeneration; however, their therapeutic efficacy requires substantial improvement. Cell priming using electrical stimulation (ES) is a promising approach to augmenting the therapeutic potential of MSCs and has shown potential for various regenerative applications. This study aimed to promote the ES-mediated chondrogenic differentiation of human MSCs and facilitate the repair of injured articular cartilage. <b>Methods:</b> MSCs were subjected to ES under various conditions (e.g., voltage, frequency, and number of repetitions) to enhance their capability of chondrogenesis and cartilage regeneration. Chondrogenic differentiation of electrically primed MSCs (epMSCs) was assessed based on gene expression and sulfated glycosaminoglycan production, and epMSCs with hyaluronic acid were transplanted into a rat osteochondral defect model. Transcriptomic analysis was performed to determine changes in gene expression by ES. <b>Results:</b> epMSCs exhibited significantly increased chondrogenic gene expression and sulfated glycosaminoglycan production compared with those in unstimulated controls. Macroscopic and histological results showed that in vivo epMSC transplantation considerably enhanced cartilage regeneration. Furthermore, ES markedly altered the expression of numerous genes of MSCs, including those associated with the extracellular matrix, the Wnt signaling pathway, and cartilage development. <b>Conclusion:</b> ES can effectively prime MSCs to improve articular cartilage repair, offering a promising strategy for enhancing the efficacy of various MSC-based therapies.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0109"},"PeriodicalIF":8.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11654951/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142857114","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 : 2024-12-18eCollection Date: 2024-01-01DOI: 10.34133/bmr.0119
Peipei Wu, Min Wang, Can Jin, Linli Li, Yuting Tang, Zhangfei Wang, Xianwen Wang, Wenrong Xu, Hui Qian
Gastric cancer (GC) is the fourth most common cancer and the second leading cause of cancer-related deaths worldwide. Despite recent advancements, clinical outcomes for GC remain unsatisfactory. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have shown promise in inhibiting tumor progression, but their role in GC, specifically human umbilical cord MSC-derived small EVs (hucMSC-sEVs), is not well understood. This study investigates the therapeutic potential of hucMSC-sEVs in GC treatment. We found that hucMSC-sEVs are captured by GC cells, substantially inhibiting their proliferation and inducing apoptosis. MiRNA sequencing revealed that hucMSC-sEVs were enriched with miRNAs having anticancer properties. Among these, miR-13896, a new miRNA, was identified as a potent inhibitor of GC cell proliferation and a promoter of apoptosis. Mechanistic studies revealed that miR-13896 targets and down-regulates the ATG2A-mediated autophagy pathway, suppressing GC cell growth and metastasis. Furthermore, we enriched hucMSC-sEVs with miR-13896 through electroporation. These engineered EVs specifically targeted tumor sites and significantly reduced GC cell growth and migration in vitro and in vivo. MiR-13896 emerged as a promising therapeutic target for GC. The delivery of miR-13896 via hucMSC-sEVs represents a novel and effective strategy for GC treatment, highlighting the potential of EV-based therapies to combat this malignancy.
{"title":"Highly Efficient Delivery of Novel MiR-13896 by Human Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Inhibits Gastric Cancer Progression by Targeting ATG2A-Mediated Autophagy.","authors":"Peipei Wu, Min Wang, Can Jin, Linli Li, Yuting Tang, Zhangfei Wang, Xianwen Wang, Wenrong Xu, Hui Qian","doi":"10.34133/bmr.0119","DOIUrl":"10.34133/bmr.0119","url":null,"abstract":"<p><p>Gastric cancer (GC) is the fourth most common cancer and the second leading cause of cancer-related deaths worldwide. Despite recent advancements, clinical outcomes for GC remain unsatisfactory. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have shown promise in inhibiting tumor progression, but their role in GC, specifically human umbilical cord MSC-derived small EVs (hucMSC-sEVs), is not well understood. This study investigates the therapeutic potential of hucMSC-sEVs in GC treatment. We found that hucMSC-sEVs are captured by GC cells, substantially inhibiting their proliferation and inducing apoptosis. MiRNA sequencing revealed that hucMSC-sEVs were enriched with miRNAs having anticancer properties. Among these, miR-13896, a new miRNA, was identified as a potent inhibitor of GC cell proliferation and a promoter of apoptosis. Mechanistic studies revealed that miR-13896 targets and down-regulates the ATG2A-mediated autophagy pathway, suppressing GC cell growth and metastasis. Furthermore, we enriched hucMSC-sEVs with miR-13896 through electroporation. These engineered EVs specifically targeted tumor sites and significantly reduced GC cell growth and migration in vitro and in vivo. MiR-13896 emerged as a promising therapeutic target for GC. The delivery of miR-13896 via hucMSC-sEVs represents a novel and effective strategy for GC treatment, highlighting the potential of EV-based therapies to combat this malignancy.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0119"},"PeriodicalIF":8.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11654722/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142857121","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 : 2024-12-11eCollection Date: 2024-01-01DOI: 10.34133/bmr.0089
Ning Zhang, Lanqing Zhao, Jinwei Li, Hongxi Li, Yu Chen
Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.
{"title":"Harnessing Nanotechnology for Gout Therapy: Colchicine-Loaded Nanoparticles Regulate Macrophage Polarization and Reduce Inflammation.","authors":"Ning Zhang, Lanqing Zhao, Jinwei Li, Hongxi Li, Yu Chen","doi":"10.34133/bmr.0089","DOIUrl":"10.34133/bmr.0089","url":null,"abstract":"<p><p>Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0089"},"PeriodicalIF":8.1,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632155/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142815260","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 : 2024-12-11eCollection Date: 2024-01-01DOI: 10.34133/bmr.0116
Yufang Gong, Kai Guo, Siyu Cai, Ke Ren, Liya Tian, Yingqi Wang, Mengyao Mu, Qingwei Meng, Jie Liu, Xiao Sun
The impact of nanoparticle size on the effectiveness of magnetic resonance imaging (MRI) using sulfurized manganese nanoparticles (MnS@PAA) stabilized with polyacrylic acid (PAA) as a binder was thoroughly investigated. MnS@PAA nanoparticles of varying sizes were synthesized by altering the ratio of ethylene glycol (EG) to diethylene glycol (DEG) during the synthesis process. These nanoparticles exhibited a uniform size distribution and demonstrated high T1 relaxation rates, along with a notable pH-responsive behavior. As the nanoparticle size increased, the T1 relaxation rate decreased, indicating that size plays a crucial role in their MRI performance. Additionally, research has revealed that the efficiency of tumor uptake by these nanoparticles is size dependent. Specifically, MnS@PAA nanoparticles with a core size of 100 nm (MS100) exhibited greater tumor accumulation and provided enhanced MRI contrast. Once within the acidic environment of a tumor, MS100 decomposes into Mn2+ and H2S. Mn2+ ions promote the generation of hydroxyl radicals, which leads to lipid peroxidation and induces ferroptosis. Concurrently, the release of H2S inhibits catalase activity, resulting in elevated levels of hydrogen peroxide (H2O2), achieving a synergistic effect between chemodynamic therapy (CDT) and gas therapy. This study explores the influence of nanoparticle size on its potential applications as an MRI contrast agent and as a therapeutic agent in cancer treatment.
{"title":"Customized Sized Manganese Sulfide Nanospheres as Efficient T<sub>1</sub> MRI Contrast Agents for Enhanced Tumor Theranostics.","authors":"Yufang Gong, Kai Guo, Siyu Cai, Ke Ren, Liya Tian, Yingqi Wang, Mengyao Mu, Qingwei Meng, Jie Liu, Xiao Sun","doi":"10.34133/bmr.0116","DOIUrl":"10.34133/bmr.0116","url":null,"abstract":"<p><p>The impact of nanoparticle size on the effectiveness of magnetic resonance imaging (MRI) using sulfurized manganese nanoparticles (MnS@PAA) stabilized with polyacrylic acid (PAA) as a binder was thoroughly investigated. MnS@PAA nanoparticles of varying sizes were synthesized by altering the ratio of ethylene glycol (EG) to diethylene glycol (DEG) during the synthesis process. These nanoparticles exhibited a uniform size distribution and demonstrated high T<sub>1</sub> relaxation rates, along with a notable pH-responsive behavior. As the nanoparticle size increased, the T<sub>1</sub> relaxation rate decreased, indicating that size plays a crucial role in their MRI performance. Additionally, research has revealed that the efficiency of tumor uptake by these nanoparticles is size dependent. Specifically, MnS@PAA nanoparticles with a core size of 100 nm (MS<sub>100</sub>) exhibited greater tumor accumulation and provided enhanced MRI contrast. Once within the acidic environment of a tumor, MS<sub>100</sub> decomposes into Mn<sup>2+</sup> and H<sub>2</sub>S. Mn<sup>2+</sup> ions promote the generation of hydroxyl radicals, which leads to lipid peroxidation and induces ferroptosis. Concurrently, the release of H<sub>2</sub>S inhibits catalase activity, resulting in elevated levels of hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>), achieving a synergistic effect between chemodynamic therapy (CDT) and gas therapy. This study explores the influence of nanoparticle size on its potential applications as an MRI contrast agent and as a therapeutic agent in cancer treatment.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0116"},"PeriodicalIF":8.1,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632153/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142815259","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 presence of anti-polyethylene glycol (anti-PEG) antibodies can hinder the therapeutic efficacy of PEGylated drugs. With the widespread use of a PEGylated coronavirus disease 2019 (COVID-19) messenger RNA vaccine (Comirnaty), the impact of pre-existing anti-PEG antibodies on vaccine potency has become a point of debate. To investigate this, we established mouse models with pre-existing anti-PEG antibodies and divided them into 3 groups: group 1 with anti-PEG immunoglobulin G + immunoglobulin M concentrations of 0.76 to 27.41 μg/ml, group 2 with concentrations of 31.27 to 99.52 μg/ml, and a naïve group with no detectable anti-PEG antibodies. Results indicated that anti-spike antibody concentrations significantly decreased in group 1 and group 2 after the 2nd vaccine dose compared to those in the naïve group. Spearman's rank correlation analysis demonstrated a negative relationship between anti-spike antibody production and anti-PEG antibody levels at both the 2nd and 3rd doses (2nd dose: ρ = -0.5296, P = 0.0031; 3rd dose: ρ = -0.387, P = 0.0381). Additionally, spike protein concentrations were 31.4-fold and 46.6-fold lower in group 1 and group 2, respectively, compared to those in the naïve group at 8 h postvaccination. The concentration of complement C3a in group 2 was significantly higher than that in the naïve group after the 3rd dose. These findings confirm that pre-existing anti-PEG antibodies diminish vaccine efficacy, alter pharmacokinetics, and elevate complement activation. Therefore, detecting pre-existing anti-PEG antibodies is crucial for optimizing vaccine efficacy, ensuring patient safety, and developing improved therapeutic strategies.
{"title":"Impact of Pre-existing Anti-polyethylene Glycol Antibodies on the Pharmacokinetics and Efficacy of a COVID-19 mRNA Vaccine (Comirnaty) In Vivo.","authors":"Yen-Ling Liu, Tzu-Yi Liao, Kai-Wen Ho, En-Shuo Liu, Bo-Cheng Huang, Shih-Ting Hong, Yuan-Chin Hsieh, Mu-Shen Chang, Bing-Tsung Wu, Fang-Ming Chen, Steve R Roffler, Chiao-Yun Chen, Yuan-Chieh Yang, Tian-Lu Cheng","doi":"10.34133/bmr.0112","DOIUrl":"10.34133/bmr.0112","url":null,"abstract":"<p><p>The presence of anti-polyethylene glycol (anti-PEG) antibodies can hinder the therapeutic efficacy of PEGylated drugs. With the widespread use of a PEGylated coronavirus disease 2019 (COVID-19) messenger RNA vaccine (Comirnaty), the impact of pre-existing anti-PEG antibodies on vaccine potency has become a point of debate. To investigate this, we established mouse models with pre-existing anti-PEG antibodies and divided them into 3 groups: group 1 with anti-PEG immunoglobulin G + immunoglobulin M concentrations of 0.76 to 27.41 μg/ml, group 2 with concentrations of 31.27 to 99.52 μg/ml, and a naïve group with no detectable anti-PEG antibodies. Results indicated that anti-spike antibody concentrations significantly decreased in group 1 and group 2 after the 2nd vaccine dose compared to those in the naïve group. Spearman's rank correlation analysis demonstrated a negative relationship between anti-spike antibody production and anti-PEG antibody levels at both the 2nd and 3rd doses (2nd dose: <i>ρ</i> = -0.5296, <i>P</i> = 0.0031; 3rd dose: <i>ρ</i> = -0.387, <i>P</i> = 0.0381). Additionally, spike protein concentrations were 31.4-fold and 46.6-fold lower in group 1 and group 2, respectively, compared to those in the naïve group at 8 h postvaccination. The concentration of complement C3a in group 2 was significantly higher than that in the naïve group after the 3rd dose. These findings confirm that pre-existing anti-PEG antibodies diminish vaccine efficacy, alter pharmacokinetics, and elevate complement activation. Therefore, detecting pre-existing anti-PEG antibodies is crucial for optimizing vaccine efficacy, ensuring patient safety, and developing improved therapeutic strategies.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0112"},"PeriodicalIF":8.1,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11633857/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142815261","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 slow rate of bone regeneration and repair in osteoporotic defects is one of the difficulties of clinical work. To prepare a novel icariin (ICA)/porous magnesium alloy scaffold and to investigate its effectiveness and possible mechanism in repairing osteoporotic bone defects, bilateral ovariectomy was performed on Sprague-Dawley rats. Then, a cylindrical bone defect was created in the model, and a novel ICA/porous magnesium alloy scaffold was prepared and implanted into the defect. Eight or 12 weeks after repairing, specimens and micro-computed tomography (CT) data were collected. Microscopic observation was fulfilled through hematoxylin and eosin, Goldner, Masson, periodic acid-Schiff, and Sirius red staining. The expression of proteins was detected by immunohistochemical staining. The novel ICA/porous magnesium alloy scaffold was noncytotoxic and biologically safe. After it was implanted into the defect for 8 or 12 weeks, the surface color and smoothness, depth, and area of the defect were better than those in the control group. Besides, there was sufficient osteoid tissue, more mineralized bones, more collagen fibers, and more polysaccharide components in the defect repaired with the ICA/porous magnesium alloy scaffold. These conditions are closer to those of real bones. Moreover, the repair effect improved with the repair time. Compared with those in the control group, the expression levels of Sirtuin 1(SIRT1), Wnt5a, β-catenin, glycogen synthase kinase 3β, alkaline phosphatase, runt-related transcription factor 2, bone morphogenetic protein-2, and osteocalcin proteins were elevated in bone tissue after the scaffold was implanted into the defect for 8 weeks (all P < 0.05). The novel ICA/porous magnesium alloy scaffold promotes the repair of osteoporotic bone defects in rats, a process that may be achieved through activation of the SIRT1-Wnt/β-catenin signaling pathway.
{"title":"Repair of Osteoporotic Bone Defects in Rats via the Sirtuin 1-Wnt/β-catenin Signaling Pathway by Novel Icariin/Porous Magnesium Alloy Scaffolds.","authors":"Fei Yu, Geng Zhang, Jian Weng, Gaozhi Jia, Chongzhou Fang, Huihui Xu, Ao Xiong, Haotian Qin, Tiantian Qi, Qi Yang, Guangyin Yuan, Hui Zeng, Yuanchao Zhu","doi":"10.34133/bmr.0090","DOIUrl":"10.34133/bmr.0090","url":null,"abstract":"<p><p>The slow rate of bone regeneration and repair in osteoporotic defects is one of the difficulties of clinical work. To prepare a novel icariin (ICA)/porous magnesium alloy scaffold and to investigate its effectiveness and possible mechanism in repairing osteoporotic bone defects, bilateral ovariectomy was performed on Sprague-Dawley rats. Then, a cylindrical bone defect was created in the model, and a novel ICA/porous magnesium alloy scaffold was prepared and implanted into the defect. Eight or 12 weeks after repairing, specimens and micro-computed tomography (CT) data were collected. Microscopic observation was fulfilled through hematoxylin and eosin, Goldner, Masson, periodic acid-Schiff, and Sirius red staining. The expression of proteins was detected by immunohistochemical staining. The novel ICA/porous magnesium alloy scaffold was noncytotoxic and biologically safe. After it was implanted into the defect for 8 or 12 weeks, the surface color and smoothness, depth, and area of the defect were better than those in the control group. Besides, there was sufficient osteoid tissue, more mineralized bones, more collagen fibers, and more polysaccharide components in the defect repaired with the ICA/porous magnesium alloy scaffold. These conditions are closer to those of real bones. Moreover, the repair effect improved with the repair time. Compared with those in the control group, the expression levels of Sirtuin 1(SIRT1), Wnt5a, β-catenin, glycogen synthase kinase 3β, alkaline phosphatase, runt-related transcription factor 2, bone morphogenetic protein-2, and osteocalcin proteins were elevated in bone tissue after the scaffold was implanted into the defect for 8 weeks (all <i>P</i> < 0.05). The novel ICA/porous magnesium alloy scaffold promotes the repair of osteoporotic bone defects in rats, a process that may be achieved through activation of the SIRT1-Wnt/β-catenin signaling pathway.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0090"},"PeriodicalIF":8.1,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11625907/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142804081","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 : 2024-12-05eCollection Date: 2024-01-01DOI: 10.34133/bmr.0115
Hohyeon Han, Sooyeon Lee, Ge Gao, Hee-Gyeong Yi, Sun Ha Paek, Jinah Jang
Chronic neuroinflammation is a principal cause of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The blood-brain barrier predominantly comprises endothelial cells, and their intercellular communication with pericytes and other cell types regulates neuroinflammation. Here, we develop a tubular, perfusable model of human cerebrovascular tissues to study neurodegenerative diseases using cerebrovascular-specific extracellular matrix bioink, derived from a complementary blend of brain- and blood-vessel-derived extracellular matrices. The endothelial cells and pericytes in the bioprinted constructs spontaneously self-assemble into a dual-layered structure, closely mimicking the anatomy of the blood-brain barrier. Moreover, the mature cerebrovascular tissue shows physiological barrier functions and neuroinflammatory responses, indicating its potential for developing models of neuroinflammation-related pathologies. Collectively, our study demonstrates that furnishing a cerebrovascular-specific microenvironment can guide the cells to have native-like anatomical relevance and functional recapitulation in vitro.
{"title":"Cerebrovascular-Specific Extracellular Matrix Bioink Promotes Blood-Brain Barrier Properties.","authors":"Hohyeon Han, Sooyeon Lee, Ge Gao, Hee-Gyeong Yi, Sun Ha Paek, Jinah Jang","doi":"10.34133/bmr.0115","DOIUrl":"10.34133/bmr.0115","url":null,"abstract":"<p><p>Chronic neuroinflammation is a principal cause of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The blood-brain barrier predominantly comprises endothelial cells, and their intercellular communication with pericytes and other cell types regulates neuroinflammation. Here, we develop a tubular, perfusable model of human cerebrovascular tissues to study neurodegenerative diseases using cerebrovascular-specific extracellular matrix bioink, derived from a complementary blend of brain- and blood-vessel-derived extracellular matrices. The endothelial cells and pericytes in the bioprinted constructs spontaneously self-assemble into a dual-layered structure, closely mimicking the anatomy of the blood-brain barrier. Moreover, the mature cerebrovascular tissue shows physiological barrier functions and neuroinflammatory responses, indicating its potential for developing models of neuroinflammation-related pathologies. Collectively, our study demonstrates that furnishing a cerebrovascular-specific microenvironment can guide the cells to have native-like anatomical relevance and functional recapitulation in vitro.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0115"},"PeriodicalIF":8.1,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11617618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142788091","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 : 2024-12-03eCollection Date: 2024-01-01DOI: 10.34133/bmr.0108
Min Liu, Siyuan Liu, Yafan Bai, Mingru Zhang, Duo Zhang, Ruijin Sun, Guyan Wang, Yulong Ma
Ischemic stroke poses a global health challenge, necessitating effective therapeutic interventions given the limited time window for thrombolytic therapy. Here, we present Res@LDH, a novel nanohybrid therapeutic agent boasting a dual reactive oxygen species scavenging efficiency of approximately 90%. Comprising Ge-containing layered double hydroxide nanosheets (Ge-LDH) as a drug nanocarrier and resveratrol as a neuroprotective agent, Res@LDH demonstrates enhanced permeability across the blood-brain barrier, ensuring high biocompatibility and stability. We explored the potential of Res@LDH in mitigating oxidative stress injury induced by middle cerebral artery occlusion and reperfusion in mice, as well as H2O2-induced cytotoxicity in HT22 cells. Our experiments unveil Res@LDH's capacity to ameliorate neurological deficits, reduce the infarction volume, mitigate blood-brain barrier disruption, exhibit a robust antioxidant activity, and dampen the release of proinflammatory cytokines. Moreover, Res@LDH treatment markedly attenuates microglial and astrocytic activation. Leveraging a pioneering synthetic approach harnessing Ge-LDH and resveratrol, Res@LDH emerges as a promising strategy for addressing ischemia-reperfusion injury, offering a concise solution to current therapeutic challenges.
{"title":"Res@LDH: A Novel Nanohybrid Therapeutic for Ischemia-Reperfusion Injury with Dual Reactive Oxygen Species Scavenging Efficiency.","authors":"Min Liu, Siyuan Liu, Yafan Bai, Mingru Zhang, Duo Zhang, Ruijin Sun, Guyan Wang, Yulong Ma","doi":"10.34133/bmr.0108","DOIUrl":"10.34133/bmr.0108","url":null,"abstract":"<p><p>Ischemic stroke poses a global health challenge, necessitating effective therapeutic interventions given the limited time window for thrombolytic therapy. Here, we present Res@LDH, a novel nanohybrid therapeutic agent boasting a dual reactive oxygen species scavenging efficiency of approximately 90%. Comprising Ge-containing layered double hydroxide nanosheets (Ge-LDH) as a drug nanocarrier and resveratrol as a neuroprotective agent, Res@LDH demonstrates enhanced permeability across the blood-brain barrier, ensuring high biocompatibility and stability. We explored the potential of Res@LDH in mitigating oxidative stress injury induced by middle cerebral artery occlusion and reperfusion in mice, as well as H<sub>2</sub>O<sub>2</sub>-induced cytotoxicity in HT22 cells. Our experiments unveil Res@LDH's capacity to ameliorate neurological deficits, reduce the infarction volume, mitigate blood-brain barrier disruption, exhibit a robust antioxidant activity, and dampen the release of proinflammatory cytokines. Moreover, Res@LDH treatment markedly attenuates microglial and astrocytic activation. Leveraging a pioneering synthetic approach harnessing Ge-LDH and resveratrol, Res@LDH emerges as a promising strategy for addressing ischemia-reperfusion injury, offering a concise solution to current therapeutic challenges.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0108"},"PeriodicalIF":8.1,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11612122/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775598","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 : 2024-11-27eCollection Date: 2024-01-01DOI: 10.34133/bmr.0117
Ding Wang, Cheng-Jie Qiu, Yaoqing Chu, Anzhuo Zhang, Ran Huang, Si-Jian Pan, Lianjiang Tan
Lung cancer remains a great threat to human health despite the rapid development of various therapeutic methods. Chemotherapy continues to be the most commonly employed treatment for lung cancer; however, it often suffers from low drug delivery efficiency and severe side effects. To enhance the therapeutic efficacy of chemotherapy, we developed a novel strategy that integrates tumor vasculature normalization with the co-delivery of therapeutic agents. This strategy employs a diblock polymeric vesicle with a reduction-sensitive linkage. Paclitaxel (PTX) is encapsulated in the bilayer, while an acid-sensitive nitric oxide (NO) precursor, DETA NONOate, and zinc oxide nanoparticles (ZnO NPs) are loaded into the central cavity. The resulting nanosystem, (ZnO,NONO)@Ves-PTX, is designed to release NO under the acidic conditions typical of the tumor microenvironment (TME) and intracellular environment. The released NO in the TME inhibits angiogenesis, thereby facilitating the delivery and distribution of therapeutic agents. Upon internalization by tumor cells, (ZnO,NONO)@Ves-PTX decomposes in response to intracellular glutathione (GSH), releasing the loaded agents. DETA NONOate and ZnO NPs generate NO and Zn2+ ions, respectively, at the intracellular pH, which synergistically inhibit tumor growth alongside PTX. This combined therapeutic approach demonstrated remarkable potential in improving the chemotherapeutic efficacy for lung cancer, offering a promising direction for future cancer treatments.
{"title":"A Polymeric Vesicle System for Combined Lung Cancer Therapy through Chemotherapy and Vasculature Normalization.","authors":"Ding Wang, Cheng-Jie Qiu, Yaoqing Chu, Anzhuo Zhang, Ran Huang, Si-Jian Pan, Lianjiang Tan","doi":"10.34133/bmr.0117","DOIUrl":"10.34133/bmr.0117","url":null,"abstract":"<p><p>Lung cancer remains a great threat to human health despite the rapid development of various therapeutic methods. Chemotherapy continues to be the most commonly employed treatment for lung cancer; however, it often suffers from low drug delivery efficiency and severe side effects. To enhance the therapeutic efficacy of chemotherapy, we developed a novel strategy that integrates tumor vasculature normalization with the co-delivery of therapeutic agents. This strategy employs a diblock polymeric vesicle with a reduction-sensitive linkage. Paclitaxel (PTX) is encapsulated in the bilayer, while an acid-sensitive nitric oxide (NO) precursor, DETA NONOate, and zinc oxide nanoparticles (ZnO NPs) are loaded into the central cavity. The resulting nanosystem, (ZnO,NONO)@Ves-PTX, is designed to release NO under the acidic conditions typical of the tumor microenvironment (TME) and intracellular environment. The released NO in the TME inhibits angiogenesis, thereby facilitating the delivery and distribution of therapeutic agents. Upon internalization by tumor cells, (ZnO,NONO)@Ves-PTX decomposes in response to intracellular glutathione (GSH), releasing the loaded agents. DETA NONOate and ZnO NPs generate NO and Zn<sup>2+</sup> ions, respectively, at the intracellular pH, which synergistically inhibit tumor growth alongside PTX. This combined therapeutic approach demonstrated remarkable potential in improving the chemotherapeutic efficacy for lung cancer, offering a promising direction for future cancer treatments.</p>","PeriodicalId":93902,"journal":{"name":"Biomaterials research","volume":"28 ","pages":"0117"},"PeriodicalIF":8.1,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11599482/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142741715","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}