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":"https://doi.org/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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","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":"https://doi.org/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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","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":"https://doi.org/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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142980212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.xpro.2024.103559
Katie M L Hanford, Kim E Dzobo, Miranda Versloot, Jorge Peter, Jeffrey Kroon
The endothelium is the gatekeeper of vessel health, and its dysfunction is pivotal in driving atherogenesis. Here, we present a protocol to replicate endothelial-macrophage crosstalk during atherogenesis, called the "atherogenesis-on-chip" model, based on the Emulate dual-channel perfusion system. We describe a model for studying endothelial-macrophage interactions during atherogenesis in human aortic endothelial cells and human macrophages using qPCR and secretome analysis, fluorescence microscopy, and flow cytometry. This protocol could be adapted toward more complex plaque microenvironment or other disease settings.
{"title":"Protocol to generate a 3D atherogenesis-on-chip model for studying endothelial-macrophage crosstalk in atherogenesis.","authors":"Katie M L Hanford, Kim E Dzobo, Miranda Versloot, Jorge Peter, Jeffrey Kroon","doi":"10.1016/j.xpro.2024.103559","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103559","url":null,"abstract":"<p><p>The endothelium is the gatekeeper of vessel health, and its dysfunction is pivotal in driving atherogenesis. Here, we present a protocol to replicate endothelial-macrophage crosstalk during atherogenesis, called the \"atherogenesis-on-chip\" model, based on the Emulate dual-channel perfusion system. We describe a model for studying endothelial-macrophage interactions during atherogenesis in human aortic endothelial cells and human macrophages using qPCR and secretome analysis, fluorescence microscopy, and flow cytometry. This protocol could be adapted toward more complex plaque microenvironment or other disease settings.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103559"},"PeriodicalIF":1.3,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972463","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.xpro.2024.103557
Stefano Conti Nibali, Andrea Magrì, Angela Messina, Armin Wagner, Ramona Duman, Vito De Pinto, Cristian Turato, Cristina Arrigoni, Marco Lolicato
Voltage-dependent anion channel 1 (VDAC1) is a key protein in cellular metabolism and apoptosis. Here, we present a protocol to express and purify milligram amounts of recombinant VDAC1 in Escherichia coli. We detail steps for a fluorescence polarization-based high-throughput screening assay using NADH displacement, along with procedures for thermostability, fluorescence polarization, and X-ray crystallography. In this context, we demonstrate how 2-methyl-2,4-pentanediol (MPD), a crystallization reagent, interferes with VDAC1 small-molecule binding, hindering the detection of these ligands in the crystal. For complete details on the use and execution of this protocol, please refer to Conti Nibali et al.1.
{"title":"Protocol for high-yield bacterial expression and purification of the voltage-dependent anion channel 1 for high-throughput biophysical assays.","authors":"Stefano Conti Nibali, Andrea Magrì, Angela Messina, Armin Wagner, Ramona Duman, Vito De Pinto, Cristian Turato, Cristina Arrigoni, Marco Lolicato","doi":"10.1016/j.xpro.2024.103557","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103557","url":null,"abstract":"<p><p>Voltage-dependent anion channel 1 (VDAC1) is a key protein in cellular metabolism and apoptosis. Here, we present a protocol to express and purify milligram amounts of recombinant VDAC1 in Escherichia coli. We detail steps for a fluorescence polarization-based high-throughput screening assay using NADH displacement, along with procedures for thermostability, fluorescence polarization, and X-ray crystallography. In this context, we demonstrate how 2-methyl-2,4-pentanediol (MPD), a crystallization reagent, interferes with VDAC1 small-molecule binding, hindering the detection of these ligands in the crystal. For complete details on the use and execution of this protocol, please refer to Conti Nibali et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103557"},"PeriodicalIF":1.3,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.xpro.2024.103545
Shaokang Mo, Kengyuan Qu, Jun Shen, Kuangyu Yen
Human pluripotent stem cells (hPSCs) provide a powerful platform for generating hematopoietic progenitor cells (HPCs) and investigating hematopoietic development. Here, we present a protocol for maintaining hPSCs and inducing their differentiation into HPCs through the endothelial-to-hematopoietic transition (EHT) on vitronectin-coated plates. We outline steps for evaluating the efficiency of HPC generation and assessing their potential to differentiate into various hematopoietic lineages. This protocol serves as a framework for exploring human hematopoiesis and generating various functional blood cells. For complete details on the use and execution of this protocol, please refer to Shen et al.1 and Qu et al.2.
{"title":"Protocol for differentiating hematopoietic progenitor cells from human pluripotent stem cells in chemically defined monolayer culture.","authors":"Shaokang Mo, Kengyuan Qu, Jun Shen, Kuangyu Yen","doi":"10.1016/j.xpro.2024.103545","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103545","url":null,"abstract":"<p><p>Human pluripotent stem cells (hPSCs) provide a powerful platform for generating hematopoietic progenitor cells (HPCs) and investigating hematopoietic development. Here, we present a protocol for maintaining hPSCs and inducing their differentiation into HPCs through the endothelial-to-hematopoietic transition (EHT) on vitronectin-coated plates. We outline steps for evaluating the efficiency of HPC generation and assessing their potential to differentiate into various hematopoietic lineages. This protocol serves as a framework for exploring human hematopoiesis and generating various functional blood cells. For complete details on the use and execution of this protocol, please refer to Shen et al.<sup>1</sup> and Qu et al.<sup>2</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103545"},"PeriodicalIF":1.3,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972428","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.xpro.2024.103566
Olga Doszyn, Tomasz Dulski, Justyna Zmorzynska
Mechanistic target of rapamycin complex 1 (mTorC1) activity plays a crucial role in brain development. Here, we present an approach for rapamycin microinjection into the habenula of larval zebrafish to achieve localized inhibition of the mTorC1 pathway and explore the role of mTorC1 in habenula function. We describe steps for performing microinjections and maintaining zebrafish larvae before and after the procedure. For complete details on the use and execution of this protocol, please refer to Doszyn et al.1.
{"title":"Protocol for microinjection of rapamycin into the zebrafish habenula.","authors":"Olga Doszyn, Tomasz Dulski, Justyna Zmorzynska","doi":"10.1016/j.xpro.2024.103566","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103566","url":null,"abstract":"<p><p>Mechanistic target of rapamycin complex 1 (mTorC1) activity plays a crucial role in brain development. Here, we present an approach for rapamycin microinjection into the habenula of larval zebrafish to achieve localized inhibition of the mTorC1 pathway and explore the role of mTorC1 in habenula function. We describe steps for performing microinjections and maintaining zebrafish larvae before and after the procedure. For complete details on the use and execution of this protocol, please refer to Doszyn et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103566"},"PeriodicalIF":1.3,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972430","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Angiogenesis begins as endothelial cells migrate, forming a sprouting tip and subsequent growth-rich stalk cells. Here, we present a protocol for transcriptomic and epigenomic analyses of tip-like cells in cultured endothelial cells. We describe steps for stimulating human umbilical vein endothelial cells (HUVECs) with vascular endothelial cell growth factor (VEGF) to generate tip-like cells. We then detail procedures for library preparation for single-cell RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq), and data analysis. This scalable protocol is also applicable to diverse omics studies, including proteomics and metabolomics. For complete details on the use and execution of this protocol, please refer to Miyamura et al.1.
{"title":"Protocol for transcriptomic and epigenomic analyses of tip-like endothelial cells using scRNA-seq and ChIP-seq.","authors":"Shintaro Funasaki, Yuri Miyamura, Shunsuke Kamei, Akhinur Rahman, Masaya Yamazaki, Shingo Usuki, Keiichiro Yasunaga, Yorifumi Satou, Hiroto Ohguchi, Takashi Minami","doi":"10.1016/j.xpro.2024.103326","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103326","url":null,"abstract":"<p><p>Angiogenesis begins as endothelial cells migrate, forming a sprouting tip and subsequent growth-rich stalk cells. Here, we present a protocol for transcriptomic and epigenomic analyses of tip-like cells in cultured endothelial cells. We describe steps for stimulating human umbilical vein endothelial cells (HUVECs) with vascular endothelial cell growth factor (VEGF) to generate tip-like cells. We then detail procedures for library preparation for single-cell RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq), and data analysis. This scalable protocol is also applicable to diverse omics studies, including proteomics and metabolomics. For complete details on the use and execution of this protocol, please refer to Miyamura et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103326"},"PeriodicalIF":1.3,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972462","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-10DOI: 10.1016/j.xpro.2024.103544
Xing-Yi Wei, Yang Pei, Li Liu, Péter Hamar, De-Sheng Pei
The recombinase polymerase amplification (RPA)-CRISPR-Cas12a-FQ system enables sensitive detection of environmental DNA (eDNA) in rare fish species. Here, we present a protocol for eDNA amplification and Cas12a for target recognition using RPA. We describe steps for identifying a target site, synthesis and purification of CRISPR RNA (crRNA), and RPA isothermal amplification. We then detail procedures for constructing the eDNA CRISPR-Cas12a detection system and verifying its sensitivity. This protocol offers a high-sensitivity approach for monitoring aquatic biodiversity and conservation efforts, even in low eDNA concentrations. For complete details on the use and execution of this protocol, please refer to Wei et al.1.
{"title":"Protocol for detecting eDNA in ecological rare fish using RPA-CRISPR-Cas12a technology.","authors":"Xing-Yi Wei, Yang Pei, Li Liu, Péter Hamar, De-Sheng Pei","doi":"10.1016/j.xpro.2024.103544","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103544","url":null,"abstract":"<p><p>The recombinase polymerase amplification (RPA)-CRISPR-Cas12a-FQ system enables sensitive detection of environmental DNA (eDNA) in rare fish species. Here, we present a protocol for eDNA amplification and Cas12a for target recognition using RPA. We describe steps for identifying a target site, synthesis and purification of CRISPR RNA (crRNA), and RPA isothermal amplification. We then detail procedures for constructing the eDNA CRISPR-Cas12a detection system and verifying its sensitivity. This protocol offers a high-sensitivity approach for monitoring aquatic biodiversity and conservation efforts, even in low eDNA concentrations. For complete details on the use and execution of this protocol, please refer to Wei et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103544"},"PeriodicalIF":1.3,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972427","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Biotinylation by antibody recognition (BAR) is an antibody-based approach for mapping proximal protein interactions in cells. Here, we present a protocol to biotinylate and identify proximal proteins using BAR. We describe steps for defining proximity labeling reaction conditions, assessing enrichment using western blot, and sample preparation for mass spectroscopy analysis. We then detail procedures for data analysis and identifying proximal proteins. This approach differs from standard proximity labeling techniques, which rely on genetically engineered enzymes fused to the target protein. For complete details on the use and execution of this protocol, please refer to Rega et al.1.
{"title":"Protocol to identify endogenous proximal proteins using biotinylation by antibody recognition.","authors":"Camilla Rega, Mercedes Pardo, Lesley-Ann Martin, Jyoti Choudhary","doi":"10.1016/j.xpro.2024.103547","DOIUrl":"https://doi.org/10.1016/j.xpro.2024.103547","url":null,"abstract":"<p><p>Biotinylation by antibody recognition (BAR) is an antibody-based approach for mapping proximal protein interactions in cells. Here, we present a protocol to biotinylate and identify proximal proteins using BAR. We describe steps for defining proximity labeling reaction conditions, assessing enrichment using western blot, and sample preparation for mass spectroscopy analysis. We then detail procedures for data analysis and identifying proximal proteins. This approach differs from standard proximity labeling techniques, which rely on genetically engineered enzymes fused to the target protein. For complete details on the use and execution of this protocol, please refer to Rega et al.<sup>1</sup>.</p>","PeriodicalId":34214,"journal":{"name":"STAR Protocols","volume":"6 1","pages":"103547"},"PeriodicalIF":1.3,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972464","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}