首页 > 最新文献

Bioactive Materials最新文献

英文 中文
Mechanically regulated microcarriers with stem cell loading for skin photoaging therapy
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-03 DOI: 10.1016/j.bioactmat.2024.12.024
Xiang Lin , Anne M. Filppula , Yuanjin Zhao , Luoran Shang , Hongbo Zhang
Long-term exposure to ultraviolet radiation compromises skin structural integrity and results in disruption of normal physiological functions. Stem cells have gained attention in anti-photoaging, while controlling the tissue mechanical microenvironment of cell delivery sites is crucial for regulating cell fate and achieving optimal therapeutic performances. Here, we introduce a mechanically regulated human recombinant collagen (RHC) microcarrier generated through microfluidics, which is capable of modulating stem cell differentiation to treat photoaged skin. By controlling the cross-linking parameters, the mechanical properties of microcarriers could precisely tuned to optimize the stem cell differentiation. The microcarriers are surface functionalized with fibronectin (Fn)-platelet derived growth factor-BB (PDGF-BB) to facilitate adipose derived mesenchymal stem cells (Ad-MSCs) loading. In in vivo experiments, subcutaneous injection of stem cell loaded RHC microcarriers significantly reduced skin wrinkles after ultraviolet-injury, effectively promoted collagen synthesis, and increased vascular density. These encouraging results indicate that the present mechanically regulated microcarriers have great potential to deliver stem cells and regulate their differentiation for anti-photoaging treatments.
{"title":"Mechanically regulated microcarriers with stem cell loading for skin photoaging therapy","authors":"Xiang Lin ,&nbsp;Anne M. Filppula ,&nbsp;Yuanjin Zhao ,&nbsp;Luoran Shang ,&nbsp;Hongbo Zhang","doi":"10.1016/j.bioactmat.2024.12.024","DOIUrl":"10.1016/j.bioactmat.2024.12.024","url":null,"abstract":"<div><div>Long-term exposure to ultraviolet radiation compromises skin structural integrity and results in disruption of normal physiological functions. Stem cells have gained attention in anti-photoaging, while controlling the tissue mechanical microenvironment of cell delivery sites is crucial for regulating cell fate and achieving optimal therapeutic performances. Here, we introduce a mechanically regulated human recombinant collagen (RHC) microcarrier generated through microfluidics, which is capable of modulating stem cell differentiation to treat photoaged skin. By controlling the cross-linking parameters, the mechanical properties of microcarriers could precisely tuned to optimize the stem cell differentiation. The microcarriers are surface functionalized with fibronectin (Fn)-platelet derived growth factor-BB (PDGF-BB) to facilitate adipose derived mesenchymal stem cells (Ad-MSCs) loading. In <em>in vivo</em> experiments, subcutaneous injection of stem cell loaded RHC microcarriers significantly reduced skin wrinkles after ultraviolet-injury, effectively promoted collagen synthesis, and increased vascular density. These encouraging results indicate that the present mechanically regulated microcarriers have great potential to deliver stem cells and regulate their differentiation for anti-photoaging treatments.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 448-456"},"PeriodicalIF":18.0,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reduce electrical overload via threaded Chinese acupuncture in nerve electrical therapy
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-03 DOI: 10.1016/j.bioactmat.2024.12.025
Yupu Liu , Yawei Du , Juan Wang , Longxi Wu, Feng Lin, Wenguo Cui
Bioelectrical stimulation is a powerful technique used to promote tissue regeneration, but it can be hindered by an “electrical overload” phenomenon in the core region of stimulation. We develop a threaded microneedle electrode system that protects against “electrical overload” by delivering medicinal hydrogel microspheres into the core regions. The threaded needle body is coated with polydopamine and chitosan to enhance the adhesion of microspheres, which are loaded into the threaded grooves, allowing for their stereoscopic release in the core regions. After the electrode is inserted, the microspheres can be delivered three-dimensionally through physical swelling and the shear-thinning effect of chitosan, mitigating the electrical damage. Microspheres are designed to release alkylated vitamin B12 and vitamin E, providing antioxidant and cell protection effects upon in-situ activation, reducing reactive oxygen species (ROS) by 72.8 % and cell death by 59.5 %. In the model of peripheral nerve injury, the electrode system improves the overall antioxidant capacity by 78.5 % and protects the surrounding cells. Additionally, it leads to an improved nerve conduction velocity ratio of 41.9 % and sciatic nerve function index of 12.1 %, indicating enhanced neuroregeneration. The threaded microneedle electrode system offers a promising approach for nerve repair by inhibiting “electrical overload”, potentially improving outcomes for tissue regeneration.
{"title":"Reduce electrical overload via threaded Chinese acupuncture in nerve electrical therapy","authors":"Yupu Liu ,&nbsp;Yawei Du ,&nbsp;Juan Wang ,&nbsp;Longxi Wu,&nbsp;Feng Lin,&nbsp;Wenguo Cui","doi":"10.1016/j.bioactmat.2024.12.025","DOIUrl":"10.1016/j.bioactmat.2024.12.025","url":null,"abstract":"<div><div>Bioelectrical stimulation is a powerful technique used to promote tissue regeneration, but it can be hindered by an “electrical overload” phenomenon in the core region of stimulation. We develop a threaded microneedle electrode system that protects against “electrical overload” by delivering medicinal hydrogel microspheres into the core regions. The threaded needle body is coated with polydopamine and chitosan to enhance the adhesion of microspheres, which are loaded into the threaded grooves, allowing for their stereoscopic release in the core regions. After the electrode is inserted, the microspheres can be delivered three-dimensionally through physical swelling and the shear-thinning effect of chitosan, mitigating the electrical damage. Microspheres are designed to release alkylated vitamin B12 and vitamin E, providing antioxidant and cell protection effects upon <em>in-situ</em> activation, reducing reactive oxygen species (ROS) by 72.8 % and cell death by 59.5 %. In the model of peripheral nerve injury, the electrode system improves the overall antioxidant capacity by 78.5 % and protects the surrounding cells. Additionally, it leads to an improved nerve conduction velocity ratio of 41.9 % and sciatic nerve function index of 12.1 %, indicating enhanced neuroregeneration. The threaded microneedle electrode system offers a promising approach for nerve repair by inhibiting “electrical overload”, potentially improving outcomes for tissue regeneration.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 476-493"},"PeriodicalIF":18.0,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754975/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multivalent ionizable lipid-polypeptides for tumor-confined mRNA transfection
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-02 DOI: 10.1016/j.bioactmat.2024.12.032
Xiaofei Zhao, Yueyue Zhang, Xin Wang, Ziming Fu, Zhiyuan Zhong, Chao Deng
mRNA therapeutics is revolutionizing the treatment concepts toward many diseases including cancer. The potential of mRNA is, however, frequently limited by modest control over site of transfection. Here, we have explored a library of multivalent ionizable lipid-polypeptides (MILP) to achieve robust mRNA complexation and tumor-confined transfection. Leveraging the multivalent electrostatic, hydrophobic, and H-bond interactions, MILP efficiently packs both mRNA and plasmid DNA into sub-80 nm nanoparticles that are stable against lyophilization and long-term storage. The best MILP@mRNA complexes afford 8-fold more cellular uptake than SM-102 lipid nanoparticle formulation (SM-102 LNP), efficient endosomal disruption, and high transfection in different cells. Interestingly, MILP@mLuc displays exclusive tumor residence and distribution via multivalency-directed strong affinity and transcytosis, and affords specific protein expression in tumor cells and macrophages at tumor sites following intratumoral injection, in sharp contrast to the indiscriminate distribution and transfection in main organs of SM-102 LNP. Notably, MILP@mIL-12 with specific and efficient cytokine expression generates significant remodeling of tumor immunoenvironments and remarkable antitumor response in subcutaneous Lewis lung carcinoma and 4T1 tumor xenografts. MILP provides a unique strategy to site-specific transfection that may greatly broaden the applications of mRNA.
{"title":"Multivalent ionizable lipid-polypeptides for tumor-confined mRNA transfection","authors":"Xiaofei Zhao,&nbsp;Yueyue Zhang,&nbsp;Xin Wang,&nbsp;Ziming Fu,&nbsp;Zhiyuan Zhong,&nbsp;Chao Deng","doi":"10.1016/j.bioactmat.2024.12.032","DOIUrl":"10.1016/j.bioactmat.2024.12.032","url":null,"abstract":"<div><div>mRNA therapeutics is revolutionizing the treatment concepts toward many diseases including cancer. The potential of mRNA is, however, frequently limited by modest control over site of transfection. Here, we have explored a library of multivalent ionizable lipid-polypeptides (MILP) to achieve robust mRNA complexation and tumor-confined transfection. Leveraging the multivalent electrostatic, hydrophobic, and H-bond interactions, MILP efficiently packs both mRNA and plasmid DNA into sub-80 nm nanoparticles that are stable against lyophilization and long-term storage. The best MILP@mRNA complexes afford 8-fold more cellular uptake than SM-102 lipid nanoparticle formulation (SM-102 LNP), efficient endosomal disruption, and high transfection in different cells. Interestingly, MILP@mLuc displays exclusive tumor residence and distribution via multivalency-directed strong affinity and transcytosis, and affords specific protein expression in tumor cells and macrophages at tumor sites following intratumoral injection, in sharp contrast to the indiscriminate distribution and transfection in main organs of SM-102 LNP. Notably, MILP@mIL-12 with specific and efficient cytokine expression generates significant remodeling of tumor immunoenvironments and remarkable antitumor response in subcutaneous Lewis lung carcinoma and 4T1 tumor xenografts. MILP provides a unique strategy to site-specific transfection that may greatly broaden the applications of mRNA.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 423-433"},"PeriodicalIF":18.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027563","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hydrogen bonding-mediated phase-transition gelatin-based bioadhesives to regulate immune microenvironment for diabetic wound healing
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-02 DOI: 10.1016/j.bioactmat.2024.12.014
Zhuoling Tian , Ruoheng Gu , Wenyue Xie , Xing Su , Zuoying Yuan , Zhuo Wan , Hao Wang , Yaqian Liu , Yuting Feng , Xiaozhi Liu , Jianyong Huang
Gelatin-based biomaterials have emerged as promising candidates for bioadhesives due to their biodegradability and biocompatibility. However, they often face limitations due to the uncontrollable phase transition of gelatin, which is dominated by hydrogen bonds between peptide chains. Here, we developed controllable phase transition gelatin-based (CPTG) bioadhesives by regulating the dynamic balance of hydrogen bonds between the peptide chains using 2-hydroxyethylurea (HU) and punicalagin (PA). These CPTG bioadhesives exhibited significant enhancements in adhesion energy and injectability even at 4 °C compared to traditional gelatin bioadhesives. The developed bioadhesives could achieve self-reinforcing interfacial adhesion upon contact with moist wound tissues. This effect was attributed to HU diffusion, which disrupted the dynamic balance of hydrogen bonds and therefore induced a localized structural densification. This process was further facilitated by the presence of pyrogallol from PA. Furthermore, the CPTG bioadhesive could modulate the immune microenvironment, offering antibacterial, antioxidant, and immune-adjustable properties, thereby accelerating diabetic wound healing, as confirmed in a diabetic wound rat model. This proposed design strategy is not only crucial for developing controllable phase-transition bioadhesives for diverse applications, but also paves the way for broadening the potential applications of gelatin-based biomaterials.
{"title":"Hydrogen bonding-mediated phase-transition gelatin-based bioadhesives to regulate immune microenvironment for diabetic wound healing","authors":"Zhuoling Tian ,&nbsp;Ruoheng Gu ,&nbsp;Wenyue Xie ,&nbsp;Xing Su ,&nbsp;Zuoying Yuan ,&nbsp;Zhuo Wan ,&nbsp;Hao Wang ,&nbsp;Yaqian Liu ,&nbsp;Yuting Feng ,&nbsp;Xiaozhi Liu ,&nbsp;Jianyong Huang","doi":"10.1016/j.bioactmat.2024.12.014","DOIUrl":"10.1016/j.bioactmat.2024.12.014","url":null,"abstract":"<div><div>Gelatin-based biomaterials have emerged as promising candidates for bioadhesives due to their biodegradability and biocompatibility. However, they often face limitations due to the uncontrollable phase transition of gelatin, which is dominated by hydrogen bonds between peptide chains. Here, we developed controllable phase transition gelatin-based (CPTG) bioadhesives by regulating the dynamic balance of hydrogen bonds between the peptide chains using 2-hydroxyethylurea (HU) and punicalagin (PA). These CPTG bioadhesives exhibited significant enhancements in adhesion energy and injectability even at 4 °C compared to traditional gelatin bioadhesives. The developed bioadhesives could achieve self-reinforcing interfacial adhesion upon contact with moist wound tissues. This effect was attributed to HU diffusion, which disrupted the dynamic balance of hydrogen bonds and therefore induced a localized structural densification. This process was further facilitated by the presence of pyrogallol from PA. Furthermore, the CPTG bioadhesive could modulate the immune microenvironment, offering antibacterial, antioxidant, and immune-adjustable properties, thereby accelerating diabetic wound healing, as confirmed in a diabetic wound rat model. This proposed design strategy is not only crucial for developing controllable phase-transition bioadhesives for diverse applications, but also paves the way for broadening the potential applications of gelatin-based biomaterials.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 434-447"},"PeriodicalIF":18.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11755075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027934","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Injectable thermosensitive antibiotic-laden chitosan hydrogel for regenerative endodontics
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-02 DOI: 10.1016/j.bioactmat.2024.12.026
Alexandre Henrique dos Reis-Prado , Maedeh Rahimnejad , Renan Dal-Fabbro , Priscila Toninatto Alves de Toledo , Caroline Anselmi , Pedro Henrique Chaves de Oliveira , J. Christopher Fenno , Luciano Tavares Angelo Cintra , Francine Benetti , Marco C. Bottino
Injectable biomaterials, such as thermosensitive chitosan (CH)-based hydrogels, present a highly translational potential in dentistry due to their minimally invasive application, adaptability to irregular defects/shapes, and ability to carry therapeutic drugs. This work explores the incorporation of azithromycin (AZI) into thermosensitive CH hydrogels for use as an intracanal medication in regenerative endodontic procedures (REPs). The morphological and chemical characteristics of the hydrogel were assessed by scanning electron microscopy (SEM), energy dispersive spectroscopy (EDS), and Fourier transform infrared spectroscopy (FTIR). The thermosensitivity, gelation kinetics, compressive strength, cytocompatibility, and antibacterial efficacy were evaluated according to well-established protocols. An in vivo model of periapical disease and evoked bleeding in rats' immature permanent teeth was performed to determine disinfection, tissue repair, and root formation. AZI was successfully incorporated into interconnected porous CH hydrogels, which retained their thermosensitivity. The mechanical and rheological findings indicated that adding AZI did not adversely affect the hydrogels’ strength and injectability. Incorporating 3 % and 5 % AZI into the hydrogels led to minimal cytotoxic effects compared to higher concentrations while enhancing the antibacterial response against endodontic bacteria. AZI-laden hydrogel significantly decreased E. faecalis biofilm compared to the controls. Regarding tissue response, the 3 % AZI-laden hydrogel improved mineralized tissue formation and vascularization compared to untreated teeth and those treated with double antibiotic paste. Our findings demonstrate that adding 3 % AZI into CH hydrogels ablates infection and supports neotissue formation in vivo when applied to a clinically relevant model of regenerative endodontics.
{"title":"Injectable thermosensitive antibiotic-laden chitosan hydrogel for regenerative endodontics","authors":"Alexandre Henrique dos Reis-Prado ,&nbsp;Maedeh Rahimnejad ,&nbsp;Renan Dal-Fabbro ,&nbsp;Priscila Toninatto Alves de Toledo ,&nbsp;Caroline Anselmi ,&nbsp;Pedro Henrique Chaves de Oliveira ,&nbsp;J. Christopher Fenno ,&nbsp;Luciano Tavares Angelo Cintra ,&nbsp;Francine Benetti ,&nbsp;Marco C. Bottino","doi":"10.1016/j.bioactmat.2024.12.026","DOIUrl":"10.1016/j.bioactmat.2024.12.026","url":null,"abstract":"<div><div>Injectable biomaterials, such as thermosensitive chitosan (CH)-based hydrogels, present a highly translational potential in dentistry due to their minimally invasive application, adaptability to irregular defects/shapes, and ability to carry therapeutic drugs. This work explores the incorporation of azithromycin (AZI) into thermosensitive CH hydrogels for use as an intracanal medication in regenerative endodontic procedures (REPs). The morphological and chemical characteristics of the hydrogel were assessed by scanning electron microscopy (SEM), energy dispersive spectroscopy (EDS), and Fourier transform infrared spectroscopy (FTIR). The thermosensitivity, gelation kinetics, compressive strength, cytocompatibility, and antibacterial efficacy were evaluated according to well-established protocols. An <em>in vivo</em> model of periapical disease and evoked bleeding in rats' immature permanent teeth was performed to determine disinfection, tissue repair, and root formation. AZI was successfully incorporated into interconnected porous CH hydrogels, which retained their thermosensitivity. The mechanical and rheological findings indicated that adding AZI did not adversely affect the hydrogels’ strength and injectability. Incorporating 3 % and 5 % AZI into the hydrogels led to minimal cytotoxic effects compared to higher concentrations while enhancing the antibacterial response against endodontic bacteria. AZI-laden hydrogel significantly decreased <em>E. faecalis</em> biofilm compared to the controls. Regarding tissue response, the 3 % AZI-laden hydrogel improved mineralized tissue formation and vascularization compared to untreated teeth and those treated with double antibiotic paste. Our findings demonstrate that adding 3 % AZI into CH hydrogels ablates infection and supports neotissue formation <em>in vivo</em> when applied to a clinically relevant model of regenerative endodontics.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 406-422"},"PeriodicalIF":18.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754974/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027937","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
bFGF-Chitosan “brain glue” promotes functional recovery after cortical ischemic stroke
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-01-02 DOI: 10.1016/j.bioactmat.2024.12.017
Jiao Mu , Xiang Zou , Xinjie Bao , Zhaoyang Yang , Peng Hao , Hongmei Duan , Wen Zhao , Yudan Gao , Jinting Wu , Kun Miao , Kwok-Fai So , Liang Chen , Ying Mao , Xiaoguang Li
The mammalian brain has an extremely limited ability to regenerate lost neurons and to recover function following ischemic stroke. A biomaterial strategy of slowly-releasing various regeneration-promoting factors to activate endogenous neurogenesis represents a safe and practical neuronal replacement therapy. In this study, basic fibroblast growth factor (bFGF)-Chitosan gel is injected into the stroke cavity. This approach promotes the proliferation of vascular endothelial cell, the formation of functional vascular network, and the final restoration of cerebral blood flow. Additionally, bFGF-Chitosan gel activates neural progenitor cells (NPCs) in the subventricular zone (SVZ), promotes the NPCs’ migration toward the stroke cavity and differentiation into mature neurons with diverse cell types (inhibitory gamma-aminobutyric acid neurons and excitatory glutamatergic neuron) and layer architecture (superficial cortex and deep cortex). These new-born neurons form functional synaptic connections with the host brain and reconstruct nascent neural networks. Furthermore, synaptogenesis in the stroke cavity and Nestin lineage cells respectively contribute to the improvement of sensorimotor function induced by bFGF-Chitosan gel after ischemic stroke. Lastly, bFGF-Chitosan gel inhibits microglia activation in the peri-infarct cortex. Our findings indicate that filling the stroke cavity with bFGF-Chitosan “brain glue” promotes angiogenesis, endogenous neurogenesis and synaptogenesis to restore function, offering innovative ideas and methods for the clinical treatment of ischemic stroke.
{"title":"bFGF-Chitosan “brain glue” promotes functional recovery after cortical ischemic stroke","authors":"Jiao Mu ,&nbsp;Xiang Zou ,&nbsp;Xinjie Bao ,&nbsp;Zhaoyang Yang ,&nbsp;Peng Hao ,&nbsp;Hongmei Duan ,&nbsp;Wen Zhao ,&nbsp;Yudan Gao ,&nbsp;Jinting Wu ,&nbsp;Kun Miao ,&nbsp;Kwok-Fai So ,&nbsp;Liang Chen ,&nbsp;Ying Mao ,&nbsp;Xiaoguang Li","doi":"10.1016/j.bioactmat.2024.12.017","DOIUrl":"10.1016/j.bioactmat.2024.12.017","url":null,"abstract":"<div><div>The mammalian brain has an extremely limited ability to regenerate lost neurons and to recover function following ischemic stroke. A biomaterial strategy of slowly-releasing various regeneration-promoting factors to activate endogenous neurogenesis represents a safe and practical neuronal replacement therapy. In this study, basic fibroblast growth factor (bFGF)-Chitosan gel is injected into the stroke cavity. This approach promotes the proliferation of vascular endothelial cell, the formation of functional vascular network, and the final restoration of cerebral blood flow. Additionally, bFGF-Chitosan gel activates neural progenitor cells (NPCs) in the subventricular zone (SVZ), promotes the NPCs’ migration toward the stroke cavity and differentiation into mature neurons with diverse cell types (inhibitory gamma-aminobutyric acid neurons and excitatory glutamatergic neuron) and layer architecture (superficial cortex and deep cortex). These new-born neurons form functional synaptic connections with the host brain and reconstruct nascent neural networks. Furthermore, synaptogenesis in the stroke cavity and Nestin lineage cells respectively contribute to the improvement of sensorimotor function induced by bFGF-Chitosan gel after ischemic stroke. Lastly, bFGF-Chitosan gel inhibits microglia activation in the peri-infarct cortex. Our findings indicate that filling the stroke cavity with bFGF-Chitosan “brain glue” promotes angiogenesis, endogenous neurogenesis and synaptogenesis to restore function, offering innovative ideas and methods for the clinical treatment of ischemic stroke.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 386-405"},"PeriodicalIF":18.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11755050/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027929","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3D bio-printed proteinaceous bioactive scaffold loaded with dual growth factor enhanced chondrogenesis and in situ cartilage regeneration
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-12-31 DOI: 10.1016/j.bioactmat.2024.12.021
Prayas Chakma Shanto , Seongsu Park , Md Abdullah Al Fahad , Myeongki Park , Byong-Taek Lee
Articular cartilage has a limited self-healing capacity, leading to joint degeneration and osteoarthritis over time. Therefore, bioactive scaffolds are gaining attention as a promising approach to regenerating and repairing damaged articular cartilage through tissue engineering. In this study, we reported on a novel 3D bio-printed proteinaceous bioactive scaffolds combined with natural porcine cancellous bone dECM, tempo-oxidized cellulose nanofiber (TOCN), and alginate carriers for TGF-β1, FGF-18, and ADSCs to repair cartilage defects. The characterization results demonstrate that the 3D scaffolds are physically stable and facilitate a controlled dual release of TGF-β1 and FGF-18. Moreover, the key biological proteins within the bioactive scaffold actively interact with the biological systems to create a favorable microenvironment for cartilage regeneration. Importantly, the in vitro, in vivo, and in silico simulation showed that the scaffolds promote stem cell recruitment, migration, proliferation, and ECM deposition, and synergistic effects of TGF-β1/FGF-18 with the bioactive scaffolds significantly regulate stem cell chondrogenesis by activating the PI3K/AKT and TGFβ1/Smad4 signaling pathways. After implantation, the proteinaceous bioactive scaffold led to the regeneration of mechanically robust, full-thickness cartilage tissue that closely resembles native cartilage. Thus, these findings may provide a promising approach for regulating stem cell chondrogenesis and treating in situ cartilage regeneration.
{"title":"3D bio-printed proteinaceous bioactive scaffold loaded with dual growth factor enhanced chondrogenesis and in situ cartilage regeneration","authors":"Prayas Chakma Shanto ,&nbsp;Seongsu Park ,&nbsp;Md Abdullah Al Fahad ,&nbsp;Myeongki Park ,&nbsp;Byong-Taek Lee","doi":"10.1016/j.bioactmat.2024.12.021","DOIUrl":"10.1016/j.bioactmat.2024.12.021","url":null,"abstract":"<div><div>Articular cartilage has a limited self-healing capacity, leading to joint degeneration and osteoarthritis over time. Therefore, bioactive scaffolds are gaining attention as a promising approach to regenerating and repairing damaged articular cartilage through tissue engineering. In this study, we reported on a novel 3D bio-printed proteinaceous bioactive scaffolds combined with natural porcine cancellous bone dECM, tempo-oxidized cellulose nanofiber (TOCN), and alginate carriers for TGF-β1, FGF-18, and ADSCs to repair cartilage defects. The characterization results demonstrate that the 3D scaffolds are physically stable and facilitate a controlled dual release of TGF-β1 and FGF-18. Moreover, the key biological proteins within the bioactive scaffold actively interact with the biological systems to create a favorable microenvironment for cartilage regeneration. Importantly, the <em>in vitro</em>, <em>in vivo</em>, and in silico simulation showed that the scaffolds promote stem cell recruitment, migration, proliferation, and ECM deposition, and synergistic effects of TGF-β1/FGF-18 with the bioactive scaffolds significantly regulate stem cell chondrogenesis by activating the PI3K/AKT and TGFβ1/Smad4 signaling pathways. After implantation, the proteinaceous bioactive scaffold led to the regeneration of mechanically robust, full-thickness cartilage tissue that closely resembles native cartilage. Thus, these findings may provide a promising approach for regulating stem cell chondrogenesis and treating in situ cartilage regeneration.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 365-385"},"PeriodicalIF":18.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11751550/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021965","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fabrication and functional validation of a hybrid biomimetic nanovaccine (HBNV) against KitK641E-mutant melanoma 抗Kit K641E突变黑色素瘤的混合仿生纳米疫苗(HBNV)的制备和功能验证
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-12-27 DOI: 10.1016/j.bioactmat.2024.12.023
Kishwor Poudel , Zhenyu Ji , Ching-Ni Njauw , Anpuchchelvi Rajadurai , Brijesh Bhayana , Ryan J. Sullivan , Jong Oh Kim , Hensin Tsao
Cancer nanovaccines hold the promise for personalization, precision, and pliability by integrating all the elements essential for effective immune stimulation. An effective immune response requires communication and interplay between antigen-presenting cells (APCs), tumor cells, and immune cells to stimulate, extend, and differentiate antigen-specific and non-specific anti-tumor immune cells. The versatility of nanomedicine can be adapted to deliver both immunoadjuvant payloads and antigens from the key players in immunity (i.e., APCs and tumor cells). The imperative for novel cancer medicine is particularly pressing for less common but more devastating KIT-mutated acral and mucosal melanomas that are resistant to small molecule c-kit and immune checkpoint inhibitors. To overcome this challenge, we successfully engineered nanotechnology-enabled hybrid biomimetic nanovaccine (HBNV) comprised of membrane proteins (antigens to activate immunity and homing/targeting ligand to tumor microenvironment (TME) and lymphoid organs) from fused cells (of APCs and tumor cells) and immunoadjuvant. These HBNVs are efficiently internalized to the target cells, assisted in the maturation of APCs via antigens and adjuvant, activated the release of anti-tumor cytokines/inhibited the release of immunosuppressive cytokine, showed a homotypic effect on TME and lymph nodes, activated the anti-tumor immune cells/downregulated the immunosuppressive immune cells, reprogram the tumor microenvironment, and showed successful anti-tumor therapeutic and prophylactic effects.
癌症纳米疫苗通过整合有效免疫刺激所需的所有要素,有望实现个性化、精准性和柔韧性。有效的免疫应答需要抗原提呈细胞(apc)、肿瘤细胞和免疫细胞之间的交流和相互作用,以刺激、扩展和分化抗原特异性和非特异性抗肿瘤免疫细胞。纳米药物的多功能性可以适应于提供免疫佐剂有效载荷和免疫关键参与者(即apc和肿瘤细胞)的抗原。对于不太常见但更具破坏性的kit突变的肢端和粘膜黑色素瘤,对小分子c-kit和免疫检查点抑制剂具有耐药性,迫切需要新的癌症药物。为了克服这一挑战,我们成功地设计了纳米技术激活的混合仿生纳米疫苗(HBNV),该疫苗由融合细胞(APCs和肿瘤细胞)和免疫佐剂的膜蛋白(激活免疫的抗原和肿瘤微环境(TME)和淋巴器官的归巢/靶向配体)组成。这些hbnv有效内化到靶细胞,通过抗原和佐剂辅助APCs成熟,激活抗肿瘤细胞因子的释放/抑制免疫抑制细胞因子的释放,对TME和淋巴结表现出同型效应,激活抗肿瘤免疫细胞/下调免疫抑制免疫细胞,重编程肿瘤微环境,并显示出成功的抗肿瘤治疗和预防作用。
{"title":"Fabrication and functional validation of a hybrid biomimetic nanovaccine (HBNV) against KitK641E-mutant melanoma","authors":"Kishwor Poudel ,&nbsp;Zhenyu Ji ,&nbsp;Ching-Ni Njauw ,&nbsp;Anpuchchelvi Rajadurai ,&nbsp;Brijesh Bhayana ,&nbsp;Ryan J. Sullivan ,&nbsp;Jong Oh Kim ,&nbsp;Hensin Tsao","doi":"10.1016/j.bioactmat.2024.12.023","DOIUrl":"10.1016/j.bioactmat.2024.12.023","url":null,"abstract":"<div><div>Cancer nanovaccines hold the promise for personalization, precision, and pliability by integrating all the elements essential for effective immune stimulation. An effective immune response requires communication and interplay between antigen-presenting cells (APCs), tumor cells, and immune cells to stimulate, extend, and differentiate antigen-specific and non-specific anti-tumor immune cells. The versatility of nanomedicine can be adapted to deliver both immunoadjuvant payloads and antigens from the key players in immunity (i.e., APCs and tumor cells). The imperative for novel cancer medicine is particularly pressing for less common but more devastating KIT-mutated acral and mucosal melanomas that are resistant to small molecule c-kit and immune checkpoint inhibitors. To overcome this challenge, we successfully engineered nanotechnology-enabled hybrid biomimetic nanovaccine (HBNV) comprised of membrane proteins (antigens to activate immunity and homing/targeting ligand to tumor microenvironment (TME) and lymphoid organs) from fused cells (of APCs and tumor cells) and immunoadjuvant. These HBNVs are efficiently internalized to the target cells, assisted in the maturation of APCs via antigens and adjuvant, activated the release of anti-tumor cytokines/inhibited the release of immunosuppressive cytokine, showed a homotypic effect on TME and lymph nodes, activated the anti-tumor immune cells/downregulated the immunosuppressive immune cells, reprogram the tumor microenvironment, and showed successful anti-tumor therapeutic and prophylactic effects.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 347-364"},"PeriodicalIF":18.0,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11742834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142999287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Degradation products of magnesium implant synergistically enhance bone regeneration: Unraveling the roles of hydrogen gas and alkaline environment 镁种植体降解产物协同促进骨再生:氢气和碱性环境作用的揭示。
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-12-26 DOI: 10.1016/j.bioactmat.2024.12.020
Yuanming An , Haozhi Zhang , Shi'an Zhang , Yuantao Zhang , Lizhen Zheng , Xin Chen , Wenxue Tong , Jiankun Xu , Ling Qin
Biodegradable magnesium (Mg) implant generally provides temporary fracture fixation and facilitates bone regeneration. However, the exact effects of generated Mg ions (Mg2+), hydrogen gas (H2), and hydroxide ions (OH-) by Mg degradation on enhancing fracture healing are not fully understood. Here we investigate the in vivo degradation of Mg intramedullary nail (Mg-IMN), revealing the generation of these degradation products around the fracture site during early stages. Bulk-RNA seq indicates that H2 and alkaline pH increase periosteal cell proliferation, while Mg2+ may mainly enhance extracellular matrix formation and cell adhesion in the femur ex vivo. In vivo studies further reveal that H2, Mg2+ and alkaline pH individually generate comparable effects to the enhanced bone regeneration in the Mg-IMN group. Mechanistically, the degradation products elevate sensory calcitonin gene-related peptide (CGRP) and simultaneously suppress adrenergic factors in newly formed bone. H2 and Mg2+, instead of alkaline pH, increase CGRP synthesis and inhibit adrenergic receptors. Our findings, for the first time, elucidate that Mg2+, H2, and alkaline pH environment generated by Mg-IMN act distinctly and synergistically mediated by the skeletal interoceptive regulation to accelerate bone regeneration. These findings may advance the understanding on biological functions of Mg-IMN in fracture repair and even other bone disorders.
可生物降解镁(Mg)种植体通常提供暂时骨折固定和促进骨再生。然而,Mg降解产生的Mg离子(Mg2+)、氢气(H2)和氢氧根离子(OH-)对促进骨折愈合的确切作用尚不完全清楚。在这里,我们研究了Mg髓内钉(Mg- imn)的体内降解,揭示了这些降解产物在早期骨折部位周围的产生。Bulk-RNA序列显示H2和碱性pH增加骨膜细胞增殖,而Mg2+可能主要增强股骨离体细胞外基质形成和细胞粘附。体内研究进一步表明,H2、Mg2+和碱性pH分别对Mg-IMN组骨再生的促进作用相当。在机制上,降解产物提高感觉降钙素基因相关肽(CGRP),同时抑制新生骨中的肾上腺素能因子。H2和Mg2+,而不是碱性pH,增加CGRP合成和抑制肾上腺素能受体。我们的研究结果首次阐明了Mg-IMN产生的Mg2+、H2和碱性pH环境通过骨骼间感受调节明显协同作用,加速骨再生。这些发现可能会促进对Mg-IMN在骨折修复甚至其他骨疾病中的生物学功能的认识。
{"title":"Degradation products of magnesium implant synergistically enhance bone regeneration: Unraveling the roles of hydrogen gas and alkaline environment","authors":"Yuanming An ,&nbsp;Haozhi Zhang ,&nbsp;Shi'an Zhang ,&nbsp;Yuantao Zhang ,&nbsp;Lizhen Zheng ,&nbsp;Xin Chen ,&nbsp;Wenxue Tong ,&nbsp;Jiankun Xu ,&nbsp;Ling Qin","doi":"10.1016/j.bioactmat.2024.12.020","DOIUrl":"10.1016/j.bioactmat.2024.12.020","url":null,"abstract":"<div><div>Biodegradable magnesium (Mg) implant generally provides temporary fracture fixation and facilitates bone regeneration. However, the exact effects of generated Mg ions (Mg<sup>2+</sup>), hydrogen gas (H<sub>2</sub>), and hydroxide ions (OH<sup>-</sup>) by Mg degradation on enhancing fracture healing are not fully understood. Here we investigate the <em>in vivo</em> degradation of Mg intramedullary nail (Mg-IMN), revealing the generation of these degradation products around the fracture site during early stages. Bulk-RNA seq indicates that H<sub>2</sub> and alkaline pH increase periosteal cell proliferation, while Mg<sup>2+</sup> may mainly enhance extracellular matrix formation and cell adhesion in the femur <em>ex vivo</em>. <em>In vivo</em> studies further reveal that H<sub>2</sub>, Mg<sup>2+</sup> and alkaline pH individually generate comparable effects to the enhanced bone regeneration in the Mg-IMN group. Mechanistically, the degradation products elevate sensory calcitonin gene-related peptide (CGRP) and simultaneously suppress adrenergic factors in newly formed bone. H<sub>2</sub> and Mg<sup>2+</sup>, instead of alkaline pH, increase CGRP synthesis and inhibit adrenergic receptors. Our findings, for the first time, elucidate that Mg<sup>2+</sup>, H<sub>2</sub>, and alkaline pH environment generated by Mg-IMN act distinctly and synergistically mediated by the skeletal interoceptive regulation to accelerate bone regeneration. These findings may advance the understanding on biological functions of Mg-IMN in fracture repair and even other bone disorders.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"46 ","pages":"Pages 331-346"},"PeriodicalIF":18.0,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732853/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142999278","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanically robust surface-degradable implant from fiber silk composites demonstrates regenerative potential 由纤维丝复合材料制成的机械坚固的表面可降解植入物具有再生潜力。
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-12-25 DOI: 10.1016/j.bioactmat.2024.11.036
Wenhan Tian , Yuzeng Liu , Bo Han , Fengqi Cheng , Kang Yang , Weiyuan Hu , Dongdong Ye , Sujun Wu , Jiping Yang , Qi Chen , Yong Hai , Robert O. Ritchie , Guanping He , Juan Guan
Through millions of years of evolution, bones have developed a complex and elegant hierarchical structure, utilizing tropocollagen and hydroxyapatite to attain an intricate balance between modulus, strength, and toughness. In this study, continuous fiber silk composites (CFSCs) of large size are prepared to mimic the hierarchical structure of natural bones, through the inheritance of the hierarchical structure of fiber silk and the integration with a polyester matrix. Due to the robust interface between the matrix and fiber silk, CFSCs show maintained stable long-term mechanical performance under wet conditions. During in vivo degradation, this material primarily undergoes host cell-mediated surface degradation, rather than bulk hydrolysis. We demonstrate significant capabilities of CFSCs in promoting vascularization and macrophage differentiation toward repair. A bone defect model further indicates the potential of CFSC for bone graft applications. Our belief is that the material family of CFSCs may promise a novel biomaterial strategy for yet to be achieved excellent regenerative implants.
经过数百万年的进化,骨骼已经发展出复杂而优雅的层次结构,利用胶原蛋白和羟基磷灰石在模量、强度和韧性之间达到复杂的平衡。在本研究中,通过继承纤维丝的层次结构并与聚酯基体结合,制备了大尺寸的连续纤维丝复合材料(CFSCs)来模拟天然骨骼的层次结构。由于基质与纤维丝之间的坚固界面,CFSCs在潮湿条件下表现出稳定的长期力学性能。在体内降解过程中,这种材料主要经历宿主细胞介导的表面降解,而不是整体水解。我们证明了CFSCs在促进血管化和巨噬细胞向修复方向分化方面的显著能力。骨缺损模型进一步表明了CFSC在骨移植应用中的潜力。我们相信,CFSCs的材料家族可能承诺一种新的生物材料策略,尚未实现优秀的再生植入物。
{"title":"Mechanically robust surface-degradable implant from fiber silk composites demonstrates regenerative potential","authors":"Wenhan Tian ,&nbsp;Yuzeng Liu ,&nbsp;Bo Han ,&nbsp;Fengqi Cheng ,&nbsp;Kang Yang ,&nbsp;Weiyuan Hu ,&nbsp;Dongdong Ye ,&nbsp;Sujun Wu ,&nbsp;Jiping Yang ,&nbsp;Qi Chen ,&nbsp;Yong Hai ,&nbsp;Robert O. Ritchie ,&nbsp;Guanping He ,&nbsp;Juan Guan","doi":"10.1016/j.bioactmat.2024.11.036","DOIUrl":"10.1016/j.bioactmat.2024.11.036","url":null,"abstract":"<div><div>Through millions of years of evolution, bones have developed a complex and elegant hierarchical structure, utilizing tropocollagen and hydroxyapatite to attain an intricate balance between modulus, strength, and toughness. In this study, continuous fiber silk composites (CFSCs) of large size are prepared to mimic the hierarchical structure of natural bones, through the inheritance of the hierarchical structure of fiber silk and the integration with a polyester matrix. Due to the robust interface between the matrix and fiber silk, CFSCs show maintained stable long-term mechanical performance under wet conditions. During <em>in vivo</em> degradation, this material primarily undergoes host cell-mediated surface degradation, rather than bulk hydrolysis. We demonstrate significant capabilities of CFSCs in promoting vascularization and macrophage differentiation toward repair. A bone defect model further indicates the potential of CFSC for bone graft applications. Our belief is that the material family of CFSCs may promise a novel biomaterial strategy for yet to be achieved excellent regenerative implants.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"45 ","pages":"Pages 584-598"},"PeriodicalIF":18.0,"publicationDate":"2024-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732114/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Bioactive Materials
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1