{"title":"Inner Ear Organoid as a Preclinical Model of Hearing Regeneration: Progress and Applications.","authors":"Yingjie Wang, Mengyu Chen, Yiyin Pan, Xianxia Li, Xiangxin Lou","doi":"10.1007/s12015-025-10941-5","DOIUrl":"https://doi.org/10.1007/s12015-025-10941-5","url":null,"abstract":"<p><p>Hearing loss is one of the most common health problems and there is no suitable way to regenerate up to now. However, if the mechanism can be clearly clarified, physiological auditory regeneration may be a possibility. Organoids, artificially generated by pluripotent stem cells under three-dimensional (3D) culture system, are now considered attractive options for constructing different 3D models to study auditory regeneration and the potential mechanisms in vitro. Organoids are essentially miniature 3D models of specific organs or tissues, enabling scientists to investigate the causes of diseases, test new drugs, and explore personalized medicine within a controlled laboratory setting. Recent discoveries have demonstrated that stem cell-derived organoids can mimic environments in vivo and provide invaluable information for modeling cochlea micro-environment. In this paper, we will provide an overview of the progress achieved in inner ear organoids as a preclinical model, aiding our understanding and providing a more effective approach to addressing hearing loss.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144708814","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}
{"title":"Chitosan-based Biomaterials in Regenerative Medicine: Optimizing Mesenchymal Stem Cell Viability and Function.","authors":"Hossein Mokhtari, Mahshid Bahari, Farshid Yeganeh","doi":"10.1007/s12015-025-10901-z","DOIUrl":"https://doi.org/10.1007/s12015-025-10901-z","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) playing a crucial role in regenerative medicine due to their multipotent differentiation capabilities and significant paracrine effects. Despite their potential, MSCs face clinical challenges, including low proliferation rates, poor survival post-transplantation, and limited tissue homing. Chitosan, a biopolymer derived from chitin, addresses these challenges effectively due to its biocompatibility, biodegradability, and ability to enhance MSC attachment, proliferation, and survival. Chitosan-based biomaterials, which can be modified through various chemical and physical methods, show substantial promise in regenerative medicine. They can be engineered into forms such as membranes, hydrogels, microgels, scaffolds, nanofibers, and nano- and microparticles and serve multiple applications from three-dimensional in vitro cultures to scaffolds for tissue engineering and in vivo cell delivery systems. Chitosan improves MSC behavior by modulating critical signaling pathways, including Wnt/β-catenin, Notch, and HIF-1α, which are essential for MSC function. Furthermore, adjusting chitosan's chemical properties can promote specific lineage differentiation and enhance MSC immunomodulatory functions, vital for therapeutic efficacy in inflammatory conditions. Currently, applications of chitosan include wound healing, which will be extended to skin regeneration, bone and cartilage repair, and vascular and neural tissue engineering. Despite progress, challenges in clinical translation persist, particularly concerning safety and standardization. Future research should aim to optimize chitosan biomaterials, refine clinical protocols, and integrate advanced technologies to enhance regenerative outcomes.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144699558","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}
Iftikhar Khan, Umama Rehman, Fatima Aslam, Saad Khan, Umna Bhatti, Hussain Ramzan, Fatima Naveed, Javeria Nawaz, Syeda Malaika Raza, Kiran Inam, Faiza Rajput, Syed Muhammad Seyab, Muhammad Riyyan, Ayesha Imran Butt, Hira Habib, Ehsanullah Alokozay
{"title":"Efficacy and Safety of Mesenchymal Stromal Cells for Steroid-Refractory Acute Graft-versus-Host Disease: An Updated Meta-Analysis of Randomized Controlled Trials.","authors":"Iftikhar Khan, Umama Rehman, Fatima Aslam, Saad Khan, Umna Bhatti, Hussain Ramzan, Fatima Naveed, Javeria Nawaz, Syeda Malaika Raza, Kiran Inam, Faiza Rajput, Syed Muhammad Seyab, Muhammad Riyyan, Ayesha Imran Butt, Hira Habib, Ehsanullah Alokozay","doi":"10.1007/s12015-025-10947-z","DOIUrl":"https://doi.org/10.1007/s12015-025-10947-z","url":null,"abstract":"<p><strong>Background: </strong>Steroid-refractory acute graft-versus-host disease (SR-aGVHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation with limited effective second-line therapies. Mesenchymal stromal cells (MSCs) have emerged as a promising therapeutic option due to their immunomodulatory properties; however, their clinical efficacy and safety remain under debate.</p><p><strong>Methods: </strong>A systematic review and meta-analysis were conducted in accordance with the PRISMA 2020 guidelines, including four randomized controlled trials (RCTs) comprising 650 patients. Primary outcomes included overall response rate (ORR), complete response (CR), and overall survival (OS). Secondary outcomes assessed organ-specific responses, adverse events (AEs), serious adverse events (SAEs), and failure-free survival.</p><p><strong>Results: </strong>MSC therapy significantly improved overall response rate (ORR) (RR = 1.13; 95% CI: 1.04-1.23; P = 0.005) and complete response (CR) rate (RR = 1.60; 95% CI: 1.35-1.90; P < 0.00001), particularly in patients with grade III-IV acute graft-versus-host disease (aGVHD) and those with gut and skin involvement. There was no significant improvement in OS (RR = 1.05; 95% CI: 0.90-1.23; P = 0.52), AEs (RR = 1.00; P = 0.96), or SAEs (RR = 1.01; P = 0.79). MSCs showed significant benefit in reducing multi-organ aGVHD (RR = 1.25; P = 0.007), but not for liver involvement or grade II-IV disease. Heterogeneity across studies was generally low to moderate.</p><p><strong>Conclusion: </strong>MSCs demonstrate significant efficacy in improving short-term clinical responses in SR-aGVHD, without increasing adverse events, particularly in cases of severe and multi-organ disease. However, they do not confer a survival benefit. Further large-scale, standardized, randomized controlled trials (RCTs) with long-term follow-up are warranted to validate these findings and inform clinical implementation.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144691552","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}
Danial Hashemi Karoii, Sobhan Bavandi, Ali Shakeri Abroudi, Melika Djamali, Hossein Azizi, Thomas Skutella
{"title":"Microarray and Single-Cell RNA Sequencing Reveals G-Protein Gene Expression Signatures of Spermatogonia Stem Cell.","authors":"Danial Hashemi Karoii, Sobhan Bavandi, Ali Shakeri Abroudi, Melika Djamali, Hossein Azizi, Thomas Skutella","doi":"10.1007/s12015-025-10942-4","DOIUrl":"https://doi.org/10.1007/s12015-025-10942-4","url":null,"abstract":"<p><p>Proper testicular development is essential for spermatogenesis, a complex biological process that depends on the continuous proliferation and differentiation of spermatogonial stem cells (SSCs). These processes are tightly regulated by the SSC niche. Understanding the developmental mechanisms of SSCs is therefore critical for elucidating the basis of male fertility. Recent studies have shown that members of the G-protein-coupled receptor (GPCR) superfamily play key roles in ion and water balance in the epididymis, development of efferent ductules, blood-epididymal barrier formation, and sperm maturation. To investigate SSC development in humans, we performed microarray analysis to examine G-protein gene expression in single cells from six human testes. Our analysis revealed that genes such as LEPROT, LRRC15, LPAR1, SSR1, BMPR2, TNFRSF11B, TNFRSF10D, DDR2, SSR3, SIGMAR1, GRIA3, OGFRL1, GRIK2, TMEM87A, GPR108, TNFRSF1A, S1PR2, and VASN were down-regulated, while FLT1, ADGRG6, CSF1R, IL7R, ADGRL3, OR4N4, MMD, SIRPB1, OR5I1, PTGDR, MPL, and GPR107 were up-regulated. Single-cell transcriptomic and bioinformatic analyses were used to validate SSC-specific gene expression and assist in SSC isolation and sorting. Additionally, immunofluorescence labeling at different developmental stages provided insights into the spatial and temporal dynamics of spermatogonia. Our findings offer new insights into the molecular mechanisms governing human SSC development and provide a valuable foundation for advancing SSC-based fertility research and therapeutic applications.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144691553","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}
Banu Çiçek Tez, Sebahat Melike Durukan, Selin Kübra Yıldır, Murat Çokkeçeci, Dudu Boyvat, Nilay Altınsoy, Fatma Fındık, Şerife Ayaz Güner, Mustafa Burak Acar, Umberto Galderisi, Servet Özcan
{"title":"Comparative Proteomic Analysis of Dental-Origin Stem Cells: Insights into Regenerative Potential.","authors":"Banu Çiçek Tez, Sebahat Melike Durukan, Selin Kübra Yıldır, Murat Çokkeçeci, Dudu Boyvat, Nilay Altınsoy, Fatma Fındık, Şerife Ayaz Güner, Mustafa Burak Acar, Umberto Galderisi, Servet Özcan","doi":"10.1007/s12015-025-10936-2","DOIUrl":"https://doi.org/10.1007/s12015-025-10936-2","url":null,"abstract":"<p><p>Teeth are a significant source of stem cells and have clinical importance for regenerative medicine. A human tooth harbors different kinds of stem cells in the dental pulp (DPSC) or the periodontal ligament (PDLSC). Also exfoliated teeth in childhood contain a special type of stem cells in their pulp called Stem cells from Human Exfoliated Deciduous teeth (SHED). All these stem cells have features and capacities that vary depending on their niche. Here we investigated the proteomic properties of three types of stem cells that originated from human teeth. We isolated and cultured the DPSCs, PDLSCs, and SHED cells. After validating MSC populations via immunophenotyping, we performed a mass spectrometry-based proteomic approach to identify and relatively quantify whole cell and secreted proteins. Identified proteins were evaluated by using Gene Ontology and Reactome pathway analysis tools. Our data reveal that SHED cells represented inflammation, hypoxia, and nutrient deficiency-associated ontologies in both their secretome and whole-cell proteomes. The whole-cell proteome of PDLSCs consisted of differentiation and proliferation-associated molecules while their secretory molecules were mainly associated with inflammation, ECM organization, and immune response. Among dental-originated stem cells, DPSCs appeared to be the healthiest and clinically relevant in terms of proteomic properties with their proliferation, growth factor signaling, and stemness-associated molecules in their secretome and whole-cell proteome. Obtained results demonstrated that every type of stem cell from dental origin has unique proteomic features that are altered by their location and physiological conditions. The findings may help researchers improve the dental stem-cell-based regenerative medicine approaches.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144675701","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}
Paul Alexis Bourgade, Napasiri Putthanbut, Jea-Young Lee, Cesario V Borlongan
{"title":"Stem Cells Run Like Clockwork for Stroke Therapeutics.","authors":"Paul Alexis Bourgade, Napasiri Putthanbut, Jea-Young Lee, Cesario V Borlongan","doi":"10.1007/s12015-025-10895-8","DOIUrl":"https://doi.org/10.1007/s12015-025-10895-8","url":null,"abstract":"","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144660279","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}
{"title":"Gene and Stem Cell-Based Therapies for Retinal Degenerative Diseases: Update, Challenges, and Future Directions.","authors":"Mohd Akbar Bhat, Shiwali Goyal","doi":"10.1007/s12015-025-10927-3","DOIUrl":"https://doi.org/10.1007/s12015-025-10927-3","url":null,"abstract":"<p><p>Retinal degenerative diseases (RDDs) are a major global cause of irreversible vision loss, primarily resulting from the progressive degeneration of photoreceptors (PRs), retinal pigment epithelium (RPE), and retinal ganglion cells (RGCs). The limited regenerative capacity of the neural retina, combined with a lack of definitive therapies, highlights the urgent need for clinically viable strategies to slow degeneration or replace lost cells. While effective clinical treatments remain unavailable, recent advances in gene and stem cell therapies offer promising avenues to restore retinal structure and function. Preclinical and clinical studies have demonstrated encouraging safety and efficacy outcomes, supporting their potential to treat both inherited and acquired forms of RDDs. Nonetheless, several challenges-including vector limitations, immune responses, and delivery constraints-continue to hinder widespread clinical adoption. This review summarizes current gene and stem cell-based therapeutic strategies, recent clinical progress, and the key challenges and future directions shaping the evolving landscape of regenerative treatment for RDDs.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144650603","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}
Claudia Alexandra Dumitru, Belal Neyazi, Tamer Ayberk Kaya, Klaus-Peter Stein, Ali Rashidi, Christian Mawrin, Ibrahim Erol Sandalcioglu
{"title":"Tissue-Resident Mesenchymal Stem/stromal Cells (MSC) Modulate the Angiogenic Processes in Brain Arteriovenous Malformations (bAVM).","authors":"Claudia Alexandra Dumitru, Belal Neyazi, Tamer Ayberk Kaya, Klaus-Peter Stein, Ali Rashidi, Christian Mawrin, Ibrahim Erol Sandalcioglu","doi":"10.1007/s12015-025-10937-1","DOIUrl":"https://doi.org/10.1007/s12015-025-10937-1","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal stem/stromal cells (MSCs) have been mainly studied in the context of cell-based therapy for a variety of medical conditions, including cerebrovascular diseases. However, the role of tissue-resident MSCs in the pathophysiology of cerebrovascular diseases in general and of brain arteriovenous malformation (bAVM) in particular is currently unknown, and was investigated in this study.</p><p><strong>Methods: </strong>Human bAVM tissues were used to identify MSCs in situ (n = 10) and to isolate them ex vivo (n = 3). The paracrine effects of bAVM-MSCs on endothelial cells (ECs) were assessed in an ex vivo model using MSC-derived supernatants (SNs) and the EC line HUVEC. Selected functional assays were validated using a second EC line (HCAEC).</p><p><strong>Results: </strong>In situ, cells with a MSC-like phenotype (CD90<sup>pos</sup>CD105<sup>pos</sup>CD73<sup>pos</sup>) were found in 7 out of 10 bAVM tissues analysed. Ex vivo, MSCs were isolated from fresh bAVM samples and were subsequently characterized according to the ISCT<sup>®</sup> criteria. The bAVM-MSC SNs had no effect on the ECs' migration, but promoted the proliferation of the ECs. The strongest stimulatory effect of the bAVM-MSC SNs was observed regarding the ECs' tubulogenesis. Additionally, the bAVM-MSC SN induced a partial endothelial-to-mesenchymal transition in ECs.</p><p><strong>Conclusions: </strong>These findings indicate that bAVMs contain tissue-resident MSCs, which can potentially modulate the biology and functions of the ECs in the bAVM microenvironment. Thus, MSCs may play critical roles in the pathophysiology and the progression of this disease.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144660280","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}
{"title":"Stem Cell for Cancer Immunotherapy: Current Approaches and Challenges.","authors":"Zainab Alali, Umme Tamanna Ferdous, Alexis Nzila, Farhana Easmin, Adnan Shakoor, Abdul Wasy Zia, Shihab Uddin","doi":"10.1007/s12015-025-10933-5","DOIUrl":"https://doi.org/10.1007/s12015-025-10933-5","url":null,"abstract":"<p><p>Stem cell-based immunotherapy represents a groundbreaking advancement in cancer treatment, leveraging the immune system's inherent capacity to target and eradicate cancer cells. This review explores some of the examples of stem cells used in cancer immunotherapy, including hematopoietic, mesenchymal, and induced pluripotent stem cells (IPSCs). It also describes stem cell functionalities like modifying tumor microenvironment (TME) and developing engineered immune cells like chimeric antigen receptor (CAR)-T cells and natural killer (NK) cells. Additionally, the clinical applications of stem cells for improving cancer immunotherapies and delivering drugs directly to solid tumors are discussed. However, several challenges limit the effectiveness of stem cell technology, including safety risks, tumor avoidance by the immune system, and regulatory protocols as well as manufacturing barriers. This article reviews current advancements to overcome these challenges, such as CRISPR-based gene editing and targeted drug delivery systems and provides an outlook on emerging trends, such as the progress of personalized stem cell therapies and the increasing effectiveness of treatment by combining them with other cancer treatments.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144620717","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}
Zhuojun Shi, Sining Chen, Wenkai Cai, Wei Chen, Yong Tang
{"title":"Advances in Photobiomodulation: Effects on Mesenchymal Stem Cells and their Paracrine Factors.","authors":"Zhuojun Shi, Sining Chen, Wenkai Cai, Wei Chen, Yong Tang","doi":"10.1007/s12015-025-10934-4","DOIUrl":"https://doi.org/10.1007/s12015-025-10934-4","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) have emerged as pivotal tools in biomedical engineering, owing to their remarkable capacity for tissue repair and regeneration. Photobiomodulation (PBM), a non-invasive and safe physical stimulation technique, has demonstrated significant potential in enhancing MSCs' cellular activity, osteogenic differentiation, and therapeutic efficacy. Despite these promising findings, several challenges hinder the clinical translation of PBM, including the optimization of irradiation parameters to maximize therapeutic outcomes and the standardization of protocols to ensure reproducibility and reliability. This review explores the current advancements in PBM technology and its application in MSC research, with a focus on understanding its mechanisms and therapeutic potential. By delving into the fine-tuning of PBM parameters, including cell factor secretion dynamics, signal transduction pathways, and cell-cell interaction networks, we aim to illuminate how PBM modulates the paracrine functions of MSCs. Additionally, the integration of PBM with biomaterials and engineering technologies presents exciting opportunities for bone tissue engineering and cell therapy. Future research should focus on uncovering the mechanisms by which PBM influences MSC behavior, optimizing its therapeutic parameters, and evaluating its safety and long-term benefits. Such efforts will pave the way for PBM's seamless integration into clinical applications, including complex bone defect repair, thereby advancing its role in precision medicine.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144601640","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}