Moataz B. Zewail, Ahmed S. Doghish, Hussein M. El-Husseiny, Eman A. Mady, Osama A. Mohammed, Abdullah M. M. Elbadry, Amir S. Elbokhomy, Abdelmenem Bhnsawy and Walaa A. El-Dakroury
{"title":"Lipid-based nanocarriers: an attractive approach for rheumatoid arthritis management","authors":"Moataz B. Zewail, Ahmed S. Doghish, Hussein M. El-Husseiny, Eman A. Mady, Osama A. Mohammed, Abdullah M. M. Elbadry, Amir S. Elbokhomy, Abdelmenem Bhnsawy and Walaa A. El-Dakroury","doi":"10.1039/D4BM01058B","DOIUrl":"10.1039/D4BM01058B","url":null,"abstract":"<p >Lipid nanoparticles (LNPs) have emerged as transformative tools in modern drug delivery, offering unparalleled potential in enhancing the efficacy and safety of various therapeutics. In the context of rheumatoid arthritis (RA), a disabling autoimmune disorder characterized by chronic inflammation, joint damage, and limited patient mobility, LNPs hold significant promise for revolutionizing treatment strategies. LNPs offer several advantages over traditional drug delivery systems, including improved pharmacokinetics, enhanced tissue penetration, and reduced systemic toxicity. This article concisely summarizes the pathogenesis of RA, its associated risk factors, and therapeutic techniques and their challenges. Additionally, it highlights the noteworthy advancements made in managing RA through LNPs, including liposomes, niosomes, bilosomes, cubosomes, spanlastics, ethosomes, solid lipid nanoparticles, lipid micelles, lipid nanocapsules, nanostructured lipid carriers, <em>etc</em>. It also delves into the specific functional attributes of these nanocarrier systems, focusing on their role in treating and monitoring RA.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6163-6195"},"PeriodicalIF":5.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142556682","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Muhammad Zubair, Saadat Hussain, Mujeeb- ur-Rehman, Ajaz Hussain, Muhammad Ehtisham Akram, Sohail Shahzad, Zahid Rauf, Maria Mujahid and Aman Ullah
{"title":"Trends in protein derived materials for wound care applications","authors":"Muhammad Zubair, Saadat Hussain, Mujeeb- ur-Rehman, Ajaz Hussain, Muhammad Ehtisham Akram, Sohail Shahzad, Zahid Rauf, Maria Mujahid and Aman Ullah","doi":"10.1039/D4BM01099J","DOIUrl":"10.1039/D4BM01099J","url":null,"abstract":"<p >Natural resource based polymers, especially those derived from proteins, have attracted significant attention for their potential utilization in advanced wound care applications. Protein based wound care materials provide superior biocompatibility, biodegradability, and other functionalities compared to conventional dressings. The effectiveness of various fabrication techniques, such as electrospinning, phase separation, self-assembly, and ball milling, is examined in the context of developing protein-based materials for wound healing. These methods produce a wide range of forms, including hydrogels, scaffolds, sponges, films, and bioinspired nanomaterials, each designed for specific types of wounds and different stages of healing. This review presents a comprehensive analysis of recent research that investigates the transformation of proteins into materials for wound healing applications. Our focus is on essential proteins, such as keratin, collagen, gelatin, silk, zein, and albumin, and we emphasize their distinct traits and roles in wound care management. Protein-based wound care materials show promising potential in biomedical engineering, offering improved healing capabilities and reduced risks of infection. It is crucial to explore the potential use of these materials in clinical settings while also addressing the challenges that may arise from their commercialization in the future.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 1","pages":" 130-160"},"PeriodicalIF":5.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142680244","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Haonan Huang, Fuxin Tang, Wenchang Gan, Ruibing Li, Zehui Hou, Taicheng Zhou and Ning Ma
{"title":"GelMA/tannic acid hydrogel decorated polypropylene mesh facilitating regeneration of abdominal wall defects†","authors":"Haonan Huang, Fuxin Tang, Wenchang Gan, Ruibing Li, Zehui Hou, Taicheng Zhou and Ning Ma","doi":"10.1039/D4BM01066C","DOIUrl":"10.1039/D4BM01066C","url":null,"abstract":"<p >Polypropylene (PP) mesh is a widely used prosthetic material in hernia repair due to its excellent mechanical properties and appropriate biocompatibility. However, its application is limited due to severe adhesion between the mesh and the abdominal viscera, leading to complications such as chronic pain, intestinal obstruction, and hernia recurrence. Currently, building anti-adhesive PP mesh remains a formidable challenge. In this work, a novel anti-adhesive PP mesh (PPM/GelMA/TA) was designed with a simple and efficient <em>in situ</em> gel of GelMA solution on the surface of PP mesh and further crosslinking of tannic acid (TA). It was demonstrated that PPM/GelMA/TA has good biocompatibility and excellent antioxidant property and effectively activates the polarization of macrophages toward the M2 phenotype <em>in vitro</em>. In addition, PPM/GelMA/TA could inhibit the growth of bacteria, which is of great significance for preventing postoperative infections. Furthermore, in the repair of full-thickness abdominal wall defects in rats, PPM/GelMA/TA reduced inflammation, promoted macrophage M2 polarization, and collagen deposition and angiogenesis so that does not cause any abdominal adhesion compared with PP mesh. As a result, our PPM/GelMA/TA shows an attractive prospect in the treatment of abdominal wall defect without adhesions.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6392-6402"},"PeriodicalIF":5.8,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hyoung-Jun Kim, Hong Man Yoon, Youngjeon Lee, Kyung Seob Lim, Jung Bae Seong, Sung-Jae Park and Yongdoo Choi
{"title":"MicBall800-coated metal clip as a novel fluorescent marker for image-guided laparoscopic surgery†","authors":"Hyoung-Jun Kim, Hong Man Yoon, Youngjeon Lee, Kyung Seob Lim, Jung Bae Seong, Sung-Jae Park and Yongdoo Choi","doi":"10.1039/D4BM01252F","DOIUrl":"10.1039/D4BM01252F","url":null,"abstract":"<p >Accurate tumor localization is crucial for the success of minimally invasive surgery, as it minimizes the resection of normal tissues surrounding tumors. Traditional methods for marking gastrointestinal (GI) tumors, such as ink tattooing, intraoperative gastroscopy or colonoscopy, and placement of metal clips, have major drawbacks in their application in laparoscopic surgery. Therefore, the development of safe and easy-to-operate marking methods for accurate and real-time detection of GI tumors during laparoscopic surgery remains an ongoing challenge. Here, we propose a new fluorescent metal clip (MicBall800 clip) for noninvasive and precise fluorescence marking of GI tumors. First, we prepared a poly(methyl methacrylate) microball with small, multiple, and separated pores in its internal structure, and near-infrared fluorescence dye (IRDye800CW) and human serum albumin complex were loaded into the pores. This near-infrared-dye-loaded poly(methyl methacrylate) microball (MicBall800) was then coated onto the surface of the metal clips to produce a highly fluorescent MicBall800 clip. Safety and biocompatibility tests of the MicBall800 clip were conducted by the Korea Testing Certification Institute. The MicBall800 clip was evaluated <em>in vivo</em> using a porcine model. The MicBall800 clip passed safety and biocompatibility tests. The MicBall800 clip could be easily marked at the target sites without causing any side effects and was detected in real time during the laparoscopic operation. The data obtained from the safety and biocompatibility tests and the <em>in vivo</em> animal study indicate that the MicBall800 clip can be an important advancement in minimally invasive and precision surgery for GI cancers.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6382-6391"},"PeriodicalIF":5.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142575477","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuhong Jiang, Lijuan Jin, Wenyu Liu, Hui Liu, Xiao Liu and Zhikai Tan
{"title":"Construction of 3D tumor in vitro models with an immune microenvironment exhibiting similar tumor properties and biomimetic physiological functionality","authors":"Yuhong Jiang, Lijuan Jin, Wenyu Liu, Hui Liu, Xiao Liu and Zhikai Tan","doi":"10.1039/D4BM00754A","DOIUrl":"10.1039/D4BM00754A","url":null,"abstract":"<p >Tumors pose a serious threat to people's lives and health, and the complex tumor microenvironment is the biggest obstacle to their treatment. In contrast to the basic protein matrices typically employed in 2D or 3D cell culture systems, decellularized extracellular matrix (dECM) can create complex microenvironments. In this study, a combination of physicochemical methods was established to obtain liver decellularized extracellular matrix scaffolds (dLECMs) to provide mechanical support and cell adhesion sites. By co-culturing tumor cells, tumor-associated stromal cells and immune cells, an <em>in vitro</em> 3D tumor model with a biomimetic immune microenvironment was constructed. By utilizing microenvironment data obtained from human liver tumor tissues and refining the double seeding modeling process, 3D <em>in vitro</em> liver tumor-like tissues with a tumor immune microenvironment (TIME) were obtained and designated as reconstructed human liver cancer (RHLC). These tissues demonstrated similar tumor characteristics and exhibited satisfactory physiological functionality. The results of metabolic characterisation and mouse tumorigenicity testing verified that the constructed RHLC significantly increased <em>in vitro</em> drug resistance while also closely mimicking <em>in vivo</em> tissue metabolism. This opens up new possibilities for creating effective <em>in vitro</em> models for screening chemotherapy drugs.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 1","pages":" 223-235"},"PeriodicalIF":5.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612897","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gege Xiong, Qiwei Chen, Qiuyu Wang, Xiaoxue Wang, Yaomu Xiao, Liuli Jin, Kaichong Yan, Xueyang Zhang and Fei Hu
{"title":"Multifaceted role of nanocomposite hydrogels in diabetic wound healing: enhanced biomedical applications and detailed molecular mechanisms","authors":"Gege Xiong, Qiwei Chen, Qiuyu Wang, Xiaoxue Wang, Yaomu Xiao, Liuli Jin, Kaichong Yan, Xueyang Zhang and Fei Hu","doi":"10.1039/D4BM01088D","DOIUrl":"10.1039/D4BM01088D","url":null,"abstract":"<p >The complex microenvironment of diabetic wounds, which is characterized by persistent hyperglycemia, excessive inflammatory responses, and hypoxic conditions, significantly impedes the efficacy of traditional hydrogels. Nanocomposite hydrogels, which combine the high-water content and biocompatibility of hydrogels with the unique functionalities of nanomaterials, offer a promising solution. These hydrogels exhibit enhanced antibacterial, antioxidant, and drug-release properties. Incorporating nanomaterials increases the mechanical strength and bioactivity of hydrogels, allowing for dynamic regulation of the wound microenvironment and promoting cell migration, proliferation, and angiogenesis, thereby accelerating wound healing. This review provides a comprehensive overview of the latest advances in nanocomposite hydrogels for diabetic wound treatment and discusses their advantages and molecular mechanisms at various healing stages. The study aims to provide a theoretical foundation and practical guidance for future research and clinical applications. Furthermore, it highlights the challenges related to the mechanical durability, antimicrobial performance, resistance issues, and interactions with the cellular environments of these hydrogels. Future directions include optimizing smart drug delivery systems and personalized medical approaches to enhance the clinical applicability of nanocomposite hydrogels.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6196-6223"},"PeriodicalIF":5.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yehua Sun, Changyang Lei, Renzhong Qiao and Chao Li
{"title":"Recent advances in carrier-free natural small molecule self-assembly for drug delivery","authors":"Yehua Sun, Changyang Lei, Renzhong Qiao and Chao Li","doi":"10.1039/D4BM01153H","DOIUrl":"10.1039/D4BM01153H","url":null,"abstract":"<p >Natural small-molecule drugs have been used for thousands of years for the prevention and treatment of human diseases. Most of the natural products available on the market have been modified into various polymer materials for improving the solubility, stability, and targeted delivery of drugs. However, these nanomedicines formed based on polymer carriers would produce severe problems such as systemic toxicity and kidney metabolic stress. In contrast, the carrier-free nanomedicines formed by their self-assembly in water have inherent advantages such as low toxicity, good biocompatibility, and biodegradability. This review summarizes the assembly process and application of natural small-molecule products, which are mainly driven by multiple non-covalent interactions, and includes single-molecule assembly, bimolecular assembly, drug-modified assembly, and organogels. Meanwhile, the molecular mechanism involved in different self-assembly processes is also discussed. Self-assembly simulation and structural modification of natural small-molecule products or traditional Chinese medicine molecules using molecular dynamics simulation and computer-assisted methods are proposed, which will lead to the discovery of more carrier-free nanomedicine drug delivery systems. Overall, this review provides an important understanding and strategy to study single-molecule and multi-molecule carrier-free nanomedicines.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6237-6252"},"PeriodicalIF":5.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sin A Park, Dajeong Hwang, Jae Hoon Kim, Seung-Yeul Lee, Jaebeom Lee, Han Sang Kim, Kyung-A Kim, Bumhee Lim, Jae-Eon Lee, Yong Hyun Jeon, Tae Jeong Oh, Jaewook Lee and Sungwhan An
{"title":"Formulation of lipid nanoparticles containing ginsenoside Rg2 and protopanaxadiol for highly efficient delivery of mRNA†","authors":"Sin A Park, Dajeong Hwang, Jae Hoon Kim, Seung-Yeul Lee, Jaebeom Lee, Han Sang Kim, Kyung-A Kim, Bumhee Lim, Jae-Eon Lee, Yong Hyun Jeon, Tae Jeong Oh, Jaewook Lee and Sungwhan An","doi":"10.1039/D4BM01070A","DOIUrl":"10.1039/D4BM01070A","url":null,"abstract":"<p >Lipid nanoparticles (LNPs) are widely recognized as crucial carriers of mRNA in therapeutic and vaccine development. The typical lipid composition of mRNA-LNP systems includes an ionizable lipid, a helper lipid, a polyethylene glycol (PEG)-lipid, and cholesterol. Concerns arise regarding cholesterol's susceptibility to oxidation, potentially leading to undesired immunological responses and toxicity. In this study, we formulated novel LNPs by replacing cholesterol with phytochemical-derived compounds, specifically ginsenoside Rg2 and its derivative phytosterol protopanaxadiol (PPD), and validated their efficacy as mRNA delivery systems. The mRNA–LNP complexes were manually prepared through a simple mixing process. The biocompatibility of these Rg2-based LNPs (Rg2-LNP) and PPD-based LNPs (PPD-LNP) was assessed through cell viability assays, while the protective function of LNPs for mRNA was demonstrated by RNase treatment. Enhanced green fluorescent protein (EGFP) mRNA delivery and expression in A549 and HeLa cells were analyzed using optical microscopy and flow cytometry. The expression efficiency of Rg2-LNP and PPD-LNP was compared with that of commercially available LNPs, with both novel formulations demonstrating superior transfection and EGFP expression. Furthermore, <em>in vivo</em> tests following intramuscular (I.M.) injection in hairless mice demonstrated efficient <em>luciferase</em> (<em>Luc</em>) mRNA delivery and effective Luc expression using Rg2-LNP and PPD-LNP compared to commercial LNPs. Results indicated that the efficiency of EGFP and Luc expression in Rg2-LNP and PPD-LNP surpassed that of the cholesterol-based LNP formulation. These findings suggest that Rg2-LNP and PPD-LNP are promising candidates for future drug and gene delivery systems.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6299-6309"},"PeriodicalIF":5.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.rsc.org/en/content/articlepdf/2024/bm/d4bm01070a?page=search","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142556681","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuhang Xu, Qian Liu, Bin Wang, Quan Li, Yue Chen, Yao Yang, Zhihao Zhu, Daohui Gong, Chuan Zhang, Guansong Wang and Hang Qian
{"title":"Correction: Tobramycin-mediated self-assembly of DNA nanostructures for targeted treatment of Pseudomonas aeruginosa-infected lung inflammation","authors":"Yuhang Xu, Qian Liu, Bin Wang, Quan Li, Yue Chen, Yao Yang, Zhihao Zhu, Daohui Gong, Chuan Zhang, Guansong Wang and Hang Qian","doi":"10.1039/D4BM90084G","DOIUrl":"10.1039/D4BM90084G","url":null,"abstract":"<p >Correction for ‘Tobramycin-mediated self-assembly of DNA nanostructures for targeted treatment of <em>Pseudomonas aeruginosa</em>-infected lung inflammation’ by Yuhang Xu <em>et al.</em>, <em>Biomater. Sci.</em>, 2024, <strong>12</strong>, 2331–2340, https://doi.org/10.1039/D3BM02121A.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 23","pages":" 6151-6151"},"PeriodicalIF":5.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.rsc.org/en/content/articlepdf/2024/bm/d4bm90084g?page=search","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142520360","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hyewon Jung, Mi-lang Kyun, Ji-In Kwon, Jeongha Kim, Ju-Kang Kim, Daeui Park, Yu Bin Lee and Kyoung-Sik Moon
{"title":"Amplified response of drug-induced liver fibrosis via immune cell co-culture in a 3D in vitro hepatic fibrosis model†","authors":"Hyewon Jung, Mi-lang Kyun, Ji-In Kwon, Jeongha Kim, Ju-Kang Kim, Daeui Park, Yu Bin Lee and Kyoung-Sik Moon","doi":"10.1039/D4BM00874J","DOIUrl":"10.1039/D4BM00874J","url":null,"abstract":"<p >Liver fibrosis, a critical consequence of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition driven by inflammation. This process involves complex interactions among hepatocytes, hepatic stellate cells (HSCs), and Kupffer cells, the liver's resident macrophages. Kupffer cells are essential in initiating fibrosis through the release of pro-inflammatory cytokines that activate HSCs. Although various <em>in vitro</em> liver fibrosis models have been developed, there is a lack of models that include the immune environment of the liver to clarify the influence of immune cells on the progression of liver fibrosis. We developed an <em>in vitro</em> liver fibrosis model by co-culturing hepatocytes (HepaRG), a hepatic stellate cell line (LX-2), and macrophages (differentiated THP-1). The effects of liver fibrosis inducers, transforming growth factor-beta1 (TGF-β1) and methotrexate (MTX), on the inflammatory response and stellate cell activation were evaluated in this triple co-culture model. A triple co-culture condition was developed as a 3D <em>in vitro</em> model using gelatin methacrylate (GelMA), offering a more biomimetic environment and achieving liver fibrosis <em>via</em> immune cell activation associated ECM deposition. In this study, the developed triple co-culture model has the potential to elucidate cell progression roles in liver fibrosis and can be applied in drug screening and toxicity assessments targeting liver fibrosis.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 24","pages":" 6351-6367"},"PeriodicalIF":5.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142556680","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}