Pub Date : 2025-01-15DOI: 10.1016/j.xpro.2024.103565
Samantha L Simpson, Ross F Collery
Ex vivo electroretinography (ERG) provides insight into the health and functionality of retinal cells, the integrity of phototransduction, and the visual cycle and allows for the direct application of pharmaceuticals to the retinal tissues. Here, we present a protocol for performing ex vivo ERG on adult zebrafish. We describe steps for zebrafish tissue dissection, mounting tissues, and assembling and connecting cassettes. We then detail procedures for running the Diagnosys software and processing and analyzing the resulting data.
{"title":"Protocol to perform ex vivo electroretinography on adult zebrafish.","authors":"Samantha L Simpson, Ross F Collery","doi":"10.1016/j.xpro.2024.103565","DOIUrl":"10.1016/j.xpro.2024.103565","url":null,"abstract":"<p><p>Ex vivo electroretinography (ERG) provides insight into the health and functionality of retinal cells, the integrity of phototransduction, and the visual cycle and allows for the direct application of pharmaceuticals to the retinal tissues. Here, we present a protocol for performing ex vivo ERG on adult zebrafish. We describe steps for zebrafish tissue dissection, mounting tissues, and assembling and connecting cassettes. We then detail procedures for running the Diagnosys software and processing and analyzing the resulting data.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103565"},"PeriodicalIF":1.3,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786734/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-15DOI: 10.1016/j.xpro.2024.103505
Katrine Ingelshed, Marit M Melssen, Diana Spiegelberg
Here, we present a protocol for guiding tissue preparation and flow cytometric analysis in subcutaneous murine tumor models and secondary lymphoid organs. We describe steps for dissociating tumors, spleens, and lymph nodes to obtain single-cell suspensions. We then detail procedures for immune cell staining and analysis and gating strategies including the use of fluorescence-minus-one controls (FMOs). This approach provides valuable insights into the impact of cancer therapies on the tumor and systemic immune response. For complete details on the use and execution of this protocol, please refer to Ingelshed et al.1.
{"title":"Protocol for in vivo immune cell analysis in subcutaneous murine tumor models using advanced flow cytometry.","authors":"Katrine Ingelshed, Marit M Melssen, Diana Spiegelberg","doi":"10.1016/j.xpro.2024.103505","DOIUrl":"10.1016/j.xpro.2024.103505","url":null,"abstract":"<p><p>Here, we present a protocol for guiding tissue preparation and flow cytometric analysis in subcutaneous murine tumor models and secondary lymphoid organs. We describe steps for dissociating tumors, spleens, and lymph nodes to obtain single-cell suspensions. We then detail procedures for immune cell staining and analysis and gating strategies including the use of fluorescence-minus-one controls (FMOs). This approach provides valuable insights into the impact of cancer therapies on the tumor and systemic immune response. For complete details on the use and execution of this protocol, please refer to Ingelshed et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103505"},"PeriodicalIF":1.3,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-15DOI: 10.1016/j.xpro.2024.103564
Jonathan D Good, Ksenia R Safina, Tyler E Miller, Peter van Galen
Single-cell RNA sequencing (scRNA-seq) enables detailed characterization of cell states but often lacks insights into tissue clonal structures. Here, we present a protocol to probe cell states and clonal information simultaneously by enriching mitochondrial DNA (mtDNA) variants from 3'-barcoded full-length cDNA. We describe steps for input library preparation, mtDNA enrichment, PCR product cleanup, and paired-end sequencing. We then detail computational steps for running maegatk, variant calling, and data integration to illuminate cell states and clonal dynamics in primary human tissues. For complete details on the use and execution of this protocol, please refer to Miller et al.1.
{"title":"Protocol for mitochondrial variant enrichment from single-cell RNA sequencing using MAESTER.","authors":"Jonathan D Good, Ksenia R Safina, Tyler E Miller, Peter van Galen","doi":"10.1016/j.xpro.2024.103564","DOIUrl":"10.1016/j.xpro.2024.103564","url":null,"abstract":"<p><p>Single-cell RNA sequencing (scRNA-seq) enables detailed characterization of cell states but often lacks insights into tissue clonal structures. Here, we present a protocol to probe cell states and clonal information simultaneously by enriching mitochondrial DNA (mtDNA) variants from 3'-barcoded full-length cDNA. We describe steps for input library preparation, mtDNA enrichment, PCR product cleanup, and paired-end sequencing. We then detail computational steps for running maegatk, variant calling, and data integration to illuminate cell states and clonal dynamics in primary human tissues. For complete details on the use and execution of this protocol, please refer to Miller et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103564"},"PeriodicalIF":1.3,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013079","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-15DOI: 10.1016/j.xpro.2024.103548
Yao Zhang, Shixiong Li, Jingyou Yu
Binding and neutralizing antibodies are critical indicators of protection against viral pathogens and are essential for assessing the immunogenicity and efficacy of a vaccine. Here, we present a protocol comprising two assays for measuring the spike-specific binding and neutralizing antibodies in mouse plasma following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. We describe steps for determining binding antibody titers using enzyme-linked immunosorbent assay (ELISA) and assessing neutralizing antibody titers through a pseudovirus neutralization assay. For complete details on the use and execution of this protocol, please refer to Jingyou Yu et al.1.
结合抗体和中和抗体是预防病毒病原体的关键指标,对评估疫苗的免疫原性和效力至关重要。在这里,我们提出了一种方案,包括两种测定方法,用于测量小鼠血浆中严重急性呼吸综合征冠状病毒2 (SARS-CoV-2)疫苗接种后的刺特异性结合抗体和中和抗体。我们描述了使用酶联免疫吸附试验(ELISA)确定结合抗体滴度和通过假病毒中和试验评估中和抗体滴度的步骤。关于本协议的使用和执行的完整细节,请参考Jingyou Yu et al.1。
{"title":"Protocol for evaluating humoral immune responses in mice following SARS-CoV-2 vaccination.","authors":"Yao Zhang, Shixiong Li, Jingyou Yu","doi":"10.1016/j.xpro.2024.103548","DOIUrl":"10.1016/j.xpro.2024.103548","url":null,"abstract":"<p><p>Binding and neutralizing antibodies are critical indicators of protection against viral pathogens and are essential for assessing the immunogenicity and efficacy of a vaccine. Here, we present a protocol comprising two assays for measuring the spike-specific binding and neutralizing antibodies in mouse plasma following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. We describe steps for determining binding antibody titers using enzyme-linked immunosorbent assay (ELISA) and assessing neutralizing antibody titers through a pseudovirus neutralization assay. For complete details on the use and execution of this protocol, please refer to Jingyou Yu et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103548"},"PeriodicalIF":1.3,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-14DOI: 10.1016/j.xpro.2024.103337
Yalan Wu, Chenguang Wang, Xiao Yu Tian
White adipose tissue (WAT) beiging holds significant therapeutic potential for combating obesity. Here, we present a protocol for inducing beige WAT in mice using both cold exposure and CL316,243 treatment. We describe steps for intraperitoneal injection, and subcutaneous WAT (sWAT) isolation, dissection, and fixation. We then detail procedures for histology, whole-mount immunofluorescence (IF) staining, and extracting RNA and protein. This protocol can be used for subsequent analysis to explore the mechanisms governing beige WAT induction in experimental settings, particularly the evaluation of angiogenesis. For complete details on the use and execution of this protocol, please refer to Wang et al.1.
{"title":"Protocol for inducing beige adipocytes in white adipose tissue of mouse using cold exposure and CL316,243 injection.","authors":"Yalan Wu, Chenguang Wang, Xiao Yu Tian","doi":"10.1016/j.xpro.2024.103337","DOIUrl":"10.1016/j.xpro.2024.103337","url":null,"abstract":"<p><p>White adipose tissue (WAT) beiging holds significant therapeutic potential for combating obesity. Here, we present a protocol for inducing beige WAT in mice using both cold exposure and CL316,243 treatment. We describe steps for intraperitoneal injection, and subcutaneous WAT (sWAT) isolation, dissection, and fixation. We then detail procedures for histology, whole-mount immunofluorescence (IF) staining, and extracting RNA and protein. This protocol can be used for subsequent analysis to explore the mechanisms governing beige WAT induction in experimental settings, particularly the evaluation of angiogenesis. For complete details on the use and execution of this protocol, please refer to Wang et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103337"},"PeriodicalIF":1.3,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11783104/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Extracellular vesicles (EVs) play a key role in cancer development and cellular homeostasis by transferring the biological cargo to recipient cells. Here, we describe steps for screening EV secretion-related genes by combining a microRNA (miRNA) library and ExoScreen, a highly sensitive EV detection technique. We also detail procedures for screening the direct target genes regulated by miRNAs. This protocol provides a useful tool for understanding complex intracellular communications involved in EV secretion. For complete details on the use and execution of this protocol, please refer to Yamamoto et al.1.
{"title":"Protocol for extracellular vesicle secretion-related gene screening via ExoScreen technique.","authors":"Tomofumi Yamamoto, Fumihiko Urabe, Yusuke Yoshioka, Yusuke Yamamoto, Takahiro Ochiya","doi":"10.1016/j.xpro.2024.103569","DOIUrl":"10.1016/j.xpro.2024.103569","url":null,"abstract":"<p><p>Extracellular vesicles (EVs) play a key role in cancer development and cellular homeostasis by transferring the biological cargo to recipient cells. Here, we describe steps for screening EV secretion-related genes by combining a microRNA (miRNA) library and ExoScreen, a highly sensitive EV detection technique. We also detail procedures for screening the direct target genes regulated by miRNAs. This protocol provides a useful tool for understanding complex intracellular communications involved in EV secretion. For complete details on the use and execution of this protocol, please refer to Yamamoto et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103569"},"PeriodicalIF":1.3,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11783109/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-13DOI: 10.1016/j.xpro.2024.103553
Martina Dzubanova, Michaela Ferencakova, Andrea Benova, Dalia Ali, Michaela Tencerova
Bone marrow stromal cells (BMSCs) serve as a valuable reservoir of multipotent stem cells important in the regulation of bone homeostasis and energy metabolism. Here, we present a protocol for isolating human BMSCs (hBMSCs) and characterizing their cellular metabolism related to hBMSC functional properties. We describe steps for bioenergetics, cell senescence, and production of reactive oxygen species (ROS), together with description of the data analysis. These assays provide information on hBMSC metabolic status valuable to regenerative medicine and therapeutic applications. For complete details on the use and execution of this protocol, please refer to Tencerova et al.1.
{"title":"Protocol for isolation of human bone marrow stromal cells and characterization of cellular metabolism.","authors":"Martina Dzubanova, Michaela Ferencakova, Andrea Benova, Dalia Ali, Michaela Tencerova","doi":"10.1016/j.xpro.2024.103553","DOIUrl":"10.1016/j.xpro.2024.103553","url":null,"abstract":"<p><p>Bone marrow stromal cells (BMSCs) serve as a valuable reservoir of multipotent stem cells important in the regulation of bone homeostasis and energy metabolism. Here, we present a protocol for isolating human BMSCs (hBMSCs) and characterizing their cellular metabolism related to hBMSC functional properties. We describe steps for bioenergetics, cell senescence, and production of reactive oxygen species (ROS), together with description of the data analysis. These assays provide information on hBMSC metabolic status valuable to regenerative medicine and therapeutic applications. For complete details on the use and execution of this protocol, please refer to Tencerova et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103553"},"PeriodicalIF":1.3,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11782812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143013076","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-12DOI: 10.1016/j.xpro.2024.103581
Yan Ma, Keshu Dong, Jie Hu, Yiyun Tang, Hanfu Xu
The silk glands (SGs) of silkworms specifically synthesize silk proteins, thus strongly influencing the yield and quality of silk. Here, we present a protocol for isolating SG nuclei from silkworms and obtaining high-quality tissue slices for spatial transcriptomics. We describe steps for rearing, dissecting, and nucleus isolation. We then detail procedures for embedding, frozen section, and RNA capturing and sequencing. This protocol enables the exploration of the spatial distribution of SG cells at single-cell resolution. For complete details on the use and execution of this protocol, please refer to Ma et al.1.
家蚕的丝腺(SG)专门合成丝蛋白,因此对丝绸的产量和质量有很大影响。在此,我们介绍了一种从蚕体内分离 SG 细胞核并获得高质量组织切片用于空间转录组学研究的方案。我们介绍了饲养、解剖和细胞核分离的步骤。然后我们详细介绍了包埋、冷冻切片、RNA捕获和测序的步骤。该方案能以单细胞分辨率探索 SG 细胞的空间分布。有关该方案使用和执行的完整细节,请参阅 Ma 等人的文章1。
{"title":"Protocol for the isolation of silk glands from silkworms for snRNA-seq and spatial transcriptomics.","authors":"Yan Ma, Keshu Dong, Jie Hu, Yiyun Tang, Hanfu Xu","doi":"10.1016/j.xpro.2024.103581","DOIUrl":"10.1016/j.xpro.2024.103581","url":null,"abstract":"<p><p>The silk glands (SGs) of silkworms specifically synthesize silk proteins, thus strongly influencing the yield and quality of silk. Here, we present a protocol for isolating SG nuclei from silkworms and obtaining high-quality tissue slices for spatial transcriptomics. We describe steps for rearing, dissecting, and nucleus isolation. We then detail procedures for embedding, frozen section, and RNA capturing and sequencing. This protocol enables the exploration of the spatial distribution of SG cells at single-cell resolution. For complete details on the use and execution of this protocol, please refer to Ma et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103581"},"PeriodicalIF":1.3,"publicationDate":"2025-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11772954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-12DOI: 10.1016/j.xpro.2024.103554
Sophia Papaioannou, Jia-Xiang See, Tinja Baljkas, Philipp Reiners-Koch, Manuel Winkler, Adelheid Cerwenka, Ana Stojanovic
Liver sinusoidal endothelial cells (LSECs) line the liver sinusoids and play a crucial role in liver function. Isolating LSECs is beneficial for their functional evaluation in vitro. Here, we provide a protocol for obtaining purified LSECs from mice via gradient centrifugation and magnetic cell sorting (MACS), yielding cells suitable for culture and downstream analyses. We describe steps for culturing the purified LSECs and demonstrate their evaluation by flow cytometry. For complete details on the use and execution of this protocol, please refer to Papaioannou et al.1.
{"title":"Protocol for isolating and purifying murine liver sinusoidal endothelial cells for in vitro culture and functional assays.","authors":"Sophia Papaioannou, Jia-Xiang See, Tinja Baljkas, Philipp Reiners-Koch, Manuel Winkler, Adelheid Cerwenka, Ana Stojanovic","doi":"10.1016/j.xpro.2024.103554","DOIUrl":"10.1016/j.xpro.2024.103554","url":null,"abstract":"<p><p>Liver sinusoidal endothelial cells (LSECs) line the liver sinusoids and play a crucial role in liver function. Isolating LSECs is beneficial for their functional evaluation in vitro. Here, we provide a protocol for obtaining purified LSECs from mice via gradient centrifugation and magnetic cell sorting (MACS), yielding cells suitable for culture and downstream analyses. We describe steps for culturing the purified LSECs and demonstrate their evaluation by flow cytometry. For complete details on the use and execution of this protocol, please refer to Papaioannou et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103554"},"PeriodicalIF":1.3,"publicationDate":"2025-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11772955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-12DOI: 10.1016/j.xpro.2024.103556
Dan Dou, Erika L F Holzbaur, C Alexander Boecker
Studies of human induced pluripotent stem cell (iPSC)-derived neurons promise important insights into neurodegenerative diseases. Here, we present a protocol for live imaging of axonal transport in glutamatergic iPSC-derived neurons (iNeurons). We describe steps for the differentiation of iPSCs into iNeurons via PiggyBac-mediated neurogenin 2 (NGN2) delivery, iNeuron culture and transfection, and the acquisition and analysis of time-lapse images. Our protocol is optimized for the widely available catalog of KOLF2.1J iPSCs with mutations relevant to neurodegenerative diseases but is also applicable to other iPSC lines. For complete details on the use and execution of this protocol, please refer to Dou et al.1,2.
{"title":"Protocol for live imaging of axonal transport in iPSC-derived iNeurons.","authors":"Dan Dou, Erika L F Holzbaur, C Alexander Boecker","doi":"10.1016/j.xpro.2024.103556","DOIUrl":"10.1016/j.xpro.2024.103556","url":null,"abstract":"<p><p>Studies of human induced pluripotent stem cell (iPSC)-derived neurons promise important insights into neurodegenerative diseases. Here, we present a protocol for live imaging of axonal transport in glutamatergic iPSC-derived neurons (iNeurons). We describe steps for the differentiation of iPSCs into iNeurons via PiggyBac-mediated neurogenin 2 (NGN2) delivery, iNeuron culture and transfection, and the acquisition and analysis of time-lapse images. Our protocol is optimized for the widely available catalog of KOLF2.1J iPSCs with mutations relevant to neurodegenerative diseases but is also applicable to other iPSC lines. For complete details on the use and execution of this protocol, please refer to Dou et al.<sup>1</sup><sup>,</sup><sup>2</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103556"},"PeriodicalIF":1.3,"publicationDate":"2025-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11772938/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}