Qiong Zhao, Jingbo Lai, Yang Jiang, Enhai Cui, Hui Chang, Ruolang Pan, Ping Li, Jian-Zhong Shao, Jing Zheng, Ye Chen
{"title":"Lactiplantibacillus plantarum -derived extracellular vesicles alleviate acute lung injury by inhibiting ferroptosis of macrophages.","authors":"Qiong Zhao, Jingbo Lai, Yang Jiang, Enhai Cui, Hui Chang, Ruolang Pan, Ping Li, Jian-Zhong Shao, Jing Zheng, Ye Chen","doi":"10.1186/s12951-025-03405-y","DOIUrl":"https://doi.org/10.1186/s12951-025-03405-y","url":null,"abstract":"<p><p>Despite considerable advancements in understanding the mechanisms of ALI, the therapeutic options available in clinical practice remain predominantly supportive, highlighting the urgent need for innovative treatments. In this study, we investigated the potential protective benefits of extracellular vehicles from the probiotic strain Lactiplantibacillus plantarum (LpEVs) in ALI mouse model. We revealed that LpEVs administration attenuated LPS-induced ALI, as evidenced by reduced lung pathology, decreased inflammatory markers, and mitigated ferroptosis. In vitro experiments demonstrated that LpEVs restrained ferroptosis and promoted a shift towards an anti-inflammatory macrophage phenotype. Moreover, LpEVs increased the expression of NRF2, resulting in the promotion of HO1 and strengthening anti-ferroptotic System Xc-/GPX4 axis. Our analysis revealed that LpEVs alleviated ALI through the suppression of macrophages ferroptosis by delivering cbn-let-7 targeting ferroptosis-related gene Acsl4. These findings propose LpEVs as a promising therapeutic approach for preventing and treating ALI, highlighting the potential of leveraging probiotic-derived biomolecules to develop novel therapeutic strategies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"307"},"PeriodicalIF":10.6,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12016285/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144024666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dongyuan Wang, Feng Yin, Zigang Li, Yu Zhang, Chen Shi
{"title":"Current progress and remaining challenges of peptide-drug conjugates (PDCs): next generation of antibody-drug conjugates (ADCs)?","authors":"Dongyuan Wang, Feng Yin, Zigang Li, Yu Zhang, Chen Shi","doi":"10.1186/s12951-025-03277-2","DOIUrl":"https://doi.org/10.1186/s12951-025-03277-2","url":null,"abstract":"<p><p>Drug conjugates have emerged as a promising alternative delivery system designed to deliver an ultra-toxic payload directly to the target cancer cells, maximizing therapeutic efficacy while minimizing toxicity. Among these, antibody-drug conjugates (ADCs) have garnered significant attention from both academia and industry due to their great potential for cancer therapy. However, peptide-drug conjugates (PDCs) offer several advantages over ADCs, including more accessible industrial synthesis, versatile functionalization, high tissue penetration, and rapid clearance with low immunotoxicity. These factors position PDCs as up-and-coming drug candidates for future cancer therapy. Despite their potential, PDCs face challenges such as poor pharmacokinetic properties and low bioactivity, which hinder their clinical development. How to design PDCs to meet clinical needs is a big challenge and urgent to resolve. In this review, we first carefully analyzed the general consideration of successful PDC design learning from ADCs. Then, we summarised the basic functions of each component of a PDC construct, comprising of peptides, linkers and payloads. The peptides in PDCs were categorized into three types: tumor targeting peptides, cell penetrating peptide and self-assembling peptide. We then analyzed the potential of these peptides for drug delivery, such as overcoming drug resistance, controlling drug release and improving therapeutic efficacy with reduced non-specific toxicity. To better understand the potential druggability of PDCs, we discussed the pharmacokinetics of PDCs and also briefly introduced the current PDCs in clinical trials. Lastly, we discussed the future perspectives for the successful development of an oncology PDC. This review aimed to provide useful information for better construction of PDCs in future clinical applications.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"305"},"PeriodicalIF":10.6,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12013038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144014618","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jayashree Nath, Goutam Banerjee, Jayita De, Noella Dsouza, Shantanu Sur, John W Scott, Pratik Banerjee
{"title":"Nanoplastics-mediated physiologic and genomic responses in pathogenic Escherichia coli O157:H7.","authors":"Jayashree Nath, Goutam Banerjee, Jayita De, Noella Dsouza, Shantanu Sur, John W Scott, Pratik Banerjee","doi":"10.1186/s12951-025-03369-z","DOIUrl":"https://doi.org/10.1186/s12951-025-03369-z","url":null,"abstract":"<p><p>The widespread occurrence of microplastics (MP) and nanoplastics (NP) in the environment is commonly thought to negatively impact living organisms; however, there remains a considerable lack of understanding regarding the actual risks associated with exposure. Microorganisms, including pathogenic bacteria, frequently interact with MPs/NPs in various ecosystems, triggering physiological responses that warrant a deeper understanding. The present study experimentally demonstrated the impact of surface-functionalized differentially charged polystyrene (PS) NPs on the physiology of human pathogenic Escherichia coli O157:H7 and their influence on biofilm formation. Our results suggest that charged NPs can influence the growth, viability, virulence, physiological stress response, and biofilm lifestyle of the pathogen. Positively-charged NPs were found to have a bacteriostatic effect on planktonic cell growth and affect cellular viability and biofilm initiation compared to negatively charged and uncharged NPs. The transcriptomic and gene expression data indicated significant changes in the global gene expression profile of cells exposed to NPs, including the differential expression of genes encoding several metabolic pathways associated with stress response and virulence. Significant upregulation of Shiga-like toxin (stx<sub>1a</sub>), quorum sensing, and biofilm initiation genes was observed in NP-exposed biofilm samples. Overall, exposure to NPs did not significantly affect the survival of pathogens but affected their growth and biofilm development pattern, and most importantly, their virulence traits.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"304"},"PeriodicalIF":10.6,"publicationDate":"2025-04-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12013119/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"PTEN as a prognostic factor for radiotherapy plus immunotherapy response in nasopharyngeal carcinoma.","authors":"Jiaxing Guo, Ming Zhang, Xiaoli Li, Jiashuo Wang","doi":"10.1186/s12951-025-03315-z","DOIUrl":"https://doi.org/10.1186/s12951-025-03315-z","url":null,"abstract":"<p><strong>Background: </strong>In the context of nasopharyngeal carcinoma (NPC) treatment, radiotherapy combined with immunotherapy (IR + RT) is gaining traction. This study focuses on analyzing exosomal proteins, particularly Phosphatase and Tensin Homolog (PTEN), for predicting the efficacy of NPC treatments. Serum samples from NPC patients and IR + RT recipients were utilized for exosome (Exo) extraction and subsequent transcriptomic and proteomic analyses to identify treatment-related proteins. Flow cytometry of cells and exosomal analysis were performed to examine these proteins. In vitro experiments using C666-1 cells and their Exos explored various cellular responses, while a murine subcutaneous NPC model investigated the impact of PTEN modulation on tumor growth and the immune microenvironment.</p><p><strong>Results: </strong>The study demonstrated that PTEN serves as a crucial predictive biomarker, with its expression changes correlated with M2 macrophage polarization and CD8+ T cell activity. This highlights the potential significance of PTEN in predicting treatment outcomes and influencing the immune response in NPC.</p><p><strong>Conclusion: </strong>The findings suggest that PTEN could play a key role in enhancing the efficacy of NPC radiotherapy and immunotherapy. By shedding light on PTEN's impact on tumor growth and the immune microenvironment, this study lays the groundwork for future personalized therapeutic strategies in NPC treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"303"},"PeriodicalIF":10.6,"publicationDate":"2025-04-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010603/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144029194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qingfu Zhang, Jianyao Su, Zhichao Li, Su Han, Chuanhe Wang, Zhijun Sun
{"title":"Migrasomes as intercellular messengers: potential in the pathological mechanism, diagnosis and treatment of clinical diseases.","authors":"Qingfu Zhang, Jianyao Su, Zhichao Li, Su Han, Chuanhe Wang, Zhijun Sun","doi":"10.1186/s12951-025-03362-6","DOIUrl":"https://doi.org/10.1186/s12951-025-03362-6","url":null,"abstract":"<p><p>Migrasomes are newly identified organelles that were first discovered in 2015. Since then, their biological structure, formation process, and physiological functions have been gradually elucidated. Research in recent years has expanded our understanding of these aspects, highlighting their significance in various physiological and pathological processes. Migrasomes have been found to play crucial roles in normal physiological functions, including embryonic development, vascular homeostasis, material transport, and mitochondrial quality control. Additionally, emerging evidence suggests their involvement in various diseases; however, clinical research on their roles remains limited. Current studies indicate that migrasomes may contribute to disease pathogenesis and hold potential for diagnostic and therapeutic applications. This review consolidates existing clinical research on migrasomes, focusing on their role in disease mechanisms and their use in medical applications. By examining their biological structure and function, this review aims to generate insights that encourage further research, ultimately contributing to advancements in disease prevention and treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"302"},"PeriodicalIF":10.6,"publicationDate":"2025-04-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12009535/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143988486","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The develop of persistent luminescence nanoparticles with excellent performances in cancer targeted bioimaging and killing: a review.","authors":"Rongshuang Tan, Jianing Wu, Chunya Wang, Zhengyan Zhao, Xiaoyuan Zhang, Chang Zhong, Zihui Tang, Rui Zheng, Binhong Du, Yunhan He, Yuhua Sun, Ping Zhou","doi":"10.1186/s12951-025-03350-w","DOIUrl":"https://doi.org/10.1186/s12951-025-03350-w","url":null,"abstract":"<p><p>The use of fluorescent nanomaterials in tumor imaging and treatment effectively avoids the original limitations of traditional tumor clinical diagnostic methods. The PLNPs emitted persistent luminescence after the end of excitation light. Owing to their superior optical properties, such as a reduced laser irradiation dose, spontaneous fluorescence interference elimination, and near-infrared imaging, PLNPs show great promise in tumor imaging. Moreover, they also achieve excellent anti-tumor therapeutic effects through surface modification and drug delivery. However, their relatively large size and limited surface modification capacity limit their ability to kill tumors effectively enough for clinical applications. Thus, this article reviews the synthesis and modification of PLNPs and the research progress in targeted tumor imaging and tumor killing. We also discuss the challenges and prospects of their future applications in these fields. This review has value for accelerating the design of PLNPs based platform for cancer diagnosis and treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"299"},"PeriodicalIF":10.6,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12007383/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022642","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Biomaterial-based drug delivery systems in the treatment of inner ear disorders.","authors":"Hong Cheng, Bin Zhang, Pei Jiang, Menghui Liao, Xin Gao, Dongyu Xu, Yusong Wang, Yangnan Hu, Huan Wang, Tingting Liu, Renjie Chai","doi":"10.1186/s12951-025-03368-0","DOIUrl":"https://doi.org/10.1186/s12951-025-03368-0","url":null,"abstract":"<p><p>Inner ear disorders are among the predominant etiology of hearing loss. The blood-labyrinth barrier limits the ability of drugs to attain pharmacologically effective concentrations within the inner ear; consequently, delivering drugs systemically is insufficient for effectively treating inner ear disorders. Hence, it is imperative to create efficient, minimal or non-invasive methods for administering drugs to the inner ear. However, the development of such a system is hindered by three main factors: anatomical unavailability, the lack of sustained drug delivery, and individual variability. Advances in biomaterials technology have created new opportunities for overcoming existing barriers, offering great hope for the effective treatment of inner ear disorders. Hydrogel- and nanoparticle-based drug delivery systems can carry drugs to targeted designated anatomical locations in the inner ear for long-term, sustained release. Furthermore, a range of devices, including microneedles, micropumps, and cochlear implants, when paired with biomaterials, enhance the delivery of drugs to the inner ear, making the treatment of inner ear disorders more effective. Therefore, biomaterial-based drug delivery systems offer the possibility for extensive clinical uses and promise to restore hearing to millions of patients with inner ear disorders.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"297"},"PeriodicalIF":10.6,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12004832/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143970298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"NIR-programmable 3D-printed shape-memory scaffold with dual-thermal responsiveness for precision bone regeneration and bone tumor management.","authors":"Hui Wang, Jiaxin Zhang, Zuhao Li, Jiaqi Liu, Haoran Chang, Shipu Jia, Zexin Di, He Liu, Jincheng Wang, Delong Gao, Chenyu Wang, Guiwei Li, Xin Zhao","doi":"10.1186/s12951-025-03375-1","DOIUrl":"https://doi.org/10.1186/s12951-025-03375-1","url":null,"abstract":"<p><p>Clinically, intraoperative treatment of bone tumors presents several challenges, including the effective inactivation of tumors and filling of irregular bone defects after tumor removal. In this study, intelligent thermosensitive composite materials with shape-memory properties were constructed using polylactic acid (PLA) and polycaprolactone (PCL), which have excellent biocompatibility and degradability. Additionally, beta-tricalcium phosphate (β-TCP), with its osteogenic properties, and magnesium (Mg) powder, with its photothermal and bone-promoting abilities, were incorporated to improve the osteogenic potential of the composite and enable the material to respond intelligently to near-infrared (NIR) light. Utilizing 3D printing technology, the composite material was prepared into an NIR-responsive shape-memory bone-filling implant that deforms when the scaffold temperature increases to 48 ℃ under NIR laser irradiation. Moreover, at a lower temperature of 42 ℃, mild photothermal therapy promotes macrophage polarization toward the M2 phenotype. This process regulates the secretion of interleukin (IL)-4, IL-10, tumor necrosis factor-α, IL-6, and bone morphogenetic protein (BMP)-2, reducing local inflammation, enhancing the release of pro-healing factors, and improving osteogenesis. Overall, this innovative scaffold is a promising and efficient treatment for filling irregular bone defects after bone tumor surgery.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"300"},"PeriodicalIF":10.6,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12007331/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143970248","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Lactobacillus acidophilus extracellular vesicles-coated UiO-66-NH<sub>2</sub>@siRNA nanoparticles for ulcerative colitis targeted gene therapy and gut microbiota modulation.","authors":"Chenyang Cui, Jiaze Tang, Jie Chen, Beining Zhang, Ruonan Li, Qiang Zhang, Chunjing Qiu, Rongchen Chen, Geng Min, Zhaowei Sun, Haibo Weng","doi":"10.1186/s12951-025-03376-0","DOIUrl":"https://doi.org/10.1186/s12951-025-03376-0","url":null,"abstract":"<p><p>Ulcerative colitis (UC) is a complex and chronic inflammatory bowel disease whose pathogenesis involves genetic and environmental factors, which poses a challenge for treatment. Here, we have designed an innovative integrated therapeutic strategy using Lactobacillus acidophilus extracellular vesicles (EVs) to encapsulate UiO-66-NH<sub>2</sub> nanoparticles bounded with TNF-α siRNA (EVs@UiO-66-NH<sub>2</sub>@siRNA) for UC treatment. This system shows superior affinity to inflammation-related cells due to the Lactobacillus acidophilus EVs can maintain immune homeostasis by regulating the secretion of cytokines in vitro. siRNA can specifically target the key inflammatory TNF-α in UC and silence its gene expression, thereby regulating the process of inflammatory response. After oral administration, EVs@UiO-66-NH<sub>2</sub>@siRNA demonstrates an accurate delivery of TNF-α siRNA to colonize the colon site and exerts a siRNA therapeutic effect by inhibiting the expression of TNF-α, which alleviates the intestinal inflammation in DSS-induced UC model. Moreover, this system can modulate the types and compositional structures of gut microbiota and metabolites to achieve an anti-inflammatory phenotype, which is helpful for the repair of intestinal homeostasis. We also have proved that UiO-66-NH<sub>2</sub> nanoparticles exhibit a high loading capacity for TNF-α siRNA and good pH responsiveness, improving the potent release of siRNA in colon tissue. Collectively, the EVs@UiO-66-NH<sub>2</sub>@siRNA nano-delivery system demonstrate a feasible combination therapeutic strategy for UC through gut microecology modulation, immune regulation and TNF-α siRNA silence, which may provide a potential targeted treatment approach for inflammatory bowel disease.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"301"},"PeriodicalIF":10.6,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12007195/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144008476","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Celery seed derived reconstituted lipid nanoparticles as an innate neuron-targeted neuroprotective nanomedicine for ischemic stroke treatment.","authors":"Dan Han, Jiaxing Zhang, Dingran Li, Cheng Wang","doi":"10.1186/s12951-025-03372-4","DOIUrl":"https://doi.org/10.1186/s12951-025-03372-4","url":null,"abstract":"<p><strong>Background: </strong>Ischemic stroke (IS) is the leading cause of worldwide death while the discovery and effective delivery of neuroprotective agents for satisfied IS treatment is still challenging.</p><p><strong>Results: </strong>In this study, we discover that celery seed (CS) derived reconstituted lipid nanoparticles (CS-rLNPs) can effectively penetrate across blood-brain barrier (BBB) with increased distribution to the brain. Especially, CS-rLNPs show innate neuron-targeting ability to primarily bind to neuron in the cerebral ischemic area, which is not reported by any parallel studies. Moreover, CS-rLNPs are found to exert therapeutic effects on IS, which effectively restore the function of model mice. Further studies reveal that the therapeutic effects are realized through TLR4/MyD88/NF-κB p65 pathway regulated anti-inflammation and anti-apoptosis mechanisms.</p><p><strong>Conclusions: </strong>Therefore, CS-rLNPs can serve as a neuron-targeted neuroprotective nanomedicine for IS treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"298"},"PeriodicalIF":10.6,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12004799/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144004531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}