EpigenomicsPub Date : 2025-10-08DOI: 10.1080/17501911.2025.2568364
Kimiya Padidar, Shaun Brennecke, John Blangero, Eric K Moses, Phillip E Melton
{"title":"Role of epigenetic markers in cardiovascular disease following hypertensive disorders of pregnancy.","authors":"Kimiya Padidar, Shaun Brennecke, John Blangero, Eric K Moses, Phillip E Melton","doi":"10.1080/17501911.2025.2568364","DOIUrl":"https://doi.org/10.1080/17501911.2025.2568364","url":null,"abstract":"<p><p>Cardiovascular disease (CVD) is the leading cause of death among women globally. Pregnancy complications, such as hypertensive disorders of pregnancy (HDP), are known to increase the risk of developing CVD. Over 10% of pregnancies globally are affected by HDP, a condition characterized by increased blood pressure and a multiorgan disorder (preeclampsia) associated with a 2- to 8-fold higher risk of hypertension, ischemic heart disease, stroke, and heart failure. Altered epigenetic regulation of angiogenesis, endothelial function, and gene expression may help explain the link between HDP and later-life CVD risk. However, studies investigating how epigenetic modifications mediate the progression from HDP to CVD remain limited. This review provides an overview on how epigenetic mechanisms may influence the long-term cardiovascular consequences of HDP. It also highlights key research gaps, including the need for long-term longitudinal studies to show causality. Further research on this topic may result in better screening, prevention strategies, and personalized therapies for women's cardiovascular health. However, epigenetic markers should be viewed as complementary to established clinical predictors, with near-term value in mechanistic risk refinement rather than as replacements for current approaches.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-10"},"PeriodicalIF":2.6,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145250461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-08DOI: 10.1080/17501911.2025.2570119
Jamaji C Nwanaji-Enwerem, Dennis Khodasevich, Andres Cardenas
{"title":"DNA methylation-predicted protein differences between Yakutian and Central Russian populations.","authors":"Jamaji C Nwanaji-Enwerem, Dennis Khodasevich, Andres Cardenas","doi":"10.1080/17501911.2025.2570119","DOIUrl":"https://doi.org/10.1080/17501911.2025.2570119","url":null,"abstract":"<p><strong>Background: </strong>Populations in subarctic regions, like Yakutia in the Russian Sakha Republic, have adapted to extreme environmental conditions, including intense cold, pronounced shifts in daylight, and variable food availability. However, the biological mechanisms underlying these adaptations remain poorly understood despite insights from genome-wide (GWAS) and epigenome-wide association studies (EWAS).</p><p><strong>Methods: </strong>Since protein profiles may more directly reflect functional physiology, we analyzed DNA methylation data from 245 healthy Russian participants using methylation-based estimators of circulating protein levels to investigate estimated proteomic differences between residents of Yakutia and Central Russia.</p><p><strong>Results: </strong>We identified regional variation in 25 protein surrogates enriched in pathways, including MET receptor activation and PI3K-Akt signaling. Some proteins mapped to previously identified GWAS genes. To our knowledge, none mapped to previously identified, differentially methylated in EWAS genes, suggesting that methylation-based protein estimation may capture distinct, complementary aspects of physiological regulation.</p><p><strong>Conclusion: </strong>These findings align with prior -omics research by highlighting regional molecular differences possibly associated with cold adaptation. They also underscore the potential of methylation-derived proteomic proxies as a useful, indirect approach for studying proteomic variation when direct protein measurements are unavailable. While promising, this method warrants further validation, particularly with respect to potential genetic confounding.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-8"},"PeriodicalIF":2.6,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145250437","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Epigenetic mechanisms in maternal-fetal crosstalk: inter- and trans-generational inheritance.","authors":"Veronica Tisato, Elisabetta D'Aversa, Francesca Salvatori, Marco Sbracia, Giuseppina Peluso, Fabio Scarpellini, Donato Gemmati","doi":"10.1080/17501911.2025.2568369","DOIUrl":"https://doi.org/10.1080/17501911.2025.2568369","url":null,"abstract":"<p><p>Epigenetics as a composite language capable of translating signals from the environment into heritable DNA modifications is revolutionizing the approach to biology and medicine. In the field of human reproductive physiology, many epigenetic marks have been decoded in the different stages of development, from gametogenesis to embryo development, pregnancy and its maintenance. These epigenetics marks are mainly ascribable to DNA methylation/demethylation processes, histone modifications, and non-coding RNAs mediated signals. Epigenetic modifiers such as DNA methyltransferases (DNMTs) and Ten-eleven translocation (TET) enzymes are involved in gene expression regulation by modifying the DNA methylation landscape, the first and best characterized epigenetic mark. In the context of human reproduction, epigenetics signals depict the mother-fetus crosstalk in which the maternal milieu dialogs with the growing embryo to establish an appropriate placental-fetal interface for a successful pregnancy. We here outline key features of the epigenetic crosstalk during the early embryo development and pregnancy establishment. The potential of inter- and trans-generational inheritance of specific epigenetic traits in response to environmental hints is also discussed. Finally, we move forward into future translational developments of epigenetics by discussing the potential use of epigenetic-based treatments (epidrugs) as upcoming therapeutic approaches in advanced personalized reproductive medicine.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-20"},"PeriodicalIF":2.6,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231902","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-06DOI: 10.1080/17501911.2025.2569301
Leonard C Steg, Isabelle M Mansuy
{"title":"A father's legacy: the sperm epigenome, preimplantation development, and paternal environment.","authors":"Leonard C Steg, Isabelle M Mansuy","doi":"10.1080/17501911.2025.2569301","DOIUrl":"https://doi.org/10.1080/17501911.2025.2569301","url":null,"abstract":"<p><p>Paternal exposure to the environment can influence offspring phenotypes via a process known as intergenerational epigenetic inheritance. Such form of inheritance involves the sperm epigenome that is subjected to modifications by paternal exposure, which are carried from the father to the next generation. After fertilization, paternally inherited changes can manifest in the embryo and result in modified phenotypes later in life. To be long-lasting, these changes must either persist, escape the epigenetic reprogramming occurring after fertilization or be reinstated by guiding mechanisms during early development. This review discusses how the sperm epigenome instructs transcription and early embryonic development, and how environmental exposure can reshape this epigenetic information to influence developmental and transcriptional programs in the embryo. It addresses the patterns of penetrance in intergenerational epigenetic inheritance and considers how the sperm and embryonic epigenome can contribute to the variability of inherited phenotypes.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-14"},"PeriodicalIF":2.6,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231908","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"SUV39H2 is a vulnerability in glioblastoma stem cells enhanced by co-targeting SUV39H1.","authors":"Bihui Cao, Qiqi Xie, Chunying Li, Jensen Mast, Bokai Wang, Qinyi Miao, Chafiq Hamdouchi, Timothy A Grese, Jia Shen","doi":"10.1080/17501911.2025.2568366","DOIUrl":"https://doi.org/10.1080/17501911.2025.2568366","url":null,"abstract":"<p><strong>Aim: </strong>To investigate the role of SUV39H2 in glioblastoma (GBM) stem cells (GSCs) and assess whether co-targeting SUV39H2 and SUV39H1 more effectively disrupts GSC maintenance, offering a potential strategy to improve GBM treatment.</p><p><strong>Methods: </strong>Single-cell RNA-seq was used to assess SUV39H2 expression in GSCs. GSC growth and stemness were evaluated using tumorsphere formation assay and extreme limiting dilution assay. Gene expression at the mRNA and protein levels was measured by RT-qPCR and western blot, respectively. Publicly available datasets were analyzed to investigate SUV39H2 expression patterns and its clinical prognostic significance in GBM and glioma.</p><p><strong>Results: </strong>SUV39H2 is overexpressed in GSCs relative to non-stem GBM cells. Its depletion impairs GSC proliferation and stemness. Co-targeting SUV39H2 and SUV39H1 enhances GSC disruption. High SUV39H2 expression correlates with poor glioma prognosis.</p><p><strong>Conclusion: </strong>SUV39H2 is a novel regulator of GSC maintenance. Dual targeting of SUV39H2 and SUV39H1 May offer a potential therapeutic approach for GBM.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-8"},"PeriodicalIF":2.6,"publicationDate":"2025-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231842","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-03DOI: 10.1080/17501911.2025.2568368
Cullen Roth, Vrinda Venu, Sasha Bacot, Shawn R Starkenburg, Christina R Steadman
{"title":"SLUR(M)-py: a SLURM powered Pythonic pipeline for parallel processing of 3D (Epi)genomic profiles.","authors":"Cullen Roth, Vrinda Venu, Sasha Bacot, Shawn R Starkenburg, Christina R Steadman","doi":"10.1080/17501911.2025.2568368","DOIUrl":"https://doi.org/10.1080/17501911.2025.2568368","url":null,"abstract":"<p><p>Epigenomics has become multi-faceted, with researchers exploring chromatin structure, nucleosome states, and epigenetic modifications, producing large, complex multi-omic datasets. Given this shift, there is a demand for bioinformatics that leverage high-performance computing (HPC) and parallelization to quickly process data. As such, we developed SLUR(M)-py, a pythonic computational platform that leverages the Simple Linux Utility for Resource Management system (SLURM) to process sequencing data. SLUR(M)-py is multi-omic and automates calls to SLURM for processing paired-end sequences from chromatin characterization experiments, including whole-genome, ChIP-seq, ATAC-seq, and Hi-C, thereby eliminating the need for multiple analytics pipelines. To demonstrate SLUR(M)-py's utility, we employ ATAC-seq and Hi-C data from viral infection experiments and the ENCODE project, and illustrate its processing speed and completeness, which outpaces current HPC pipelines. We explore the effect of dropping duplicate sequenced reads in ATAC-seq, demonstrate how SLUR(M)-py can be used for quality control, and how to detect artifacts in Hi-C from viral infection experiments. Finally, we show how features in SLUR(M)-py, like inter-chromosomal analysis, can be used to explore the dynamics of chromosomal contacts in mammalian cells. This multi-omic, system-agnostic platform eases the computational burden for researchers and quickly produces accurate and reliable data analytics for the epigenomics community.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-15"},"PeriodicalIF":2.6,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-02DOI: 10.1080/17501911.2025.2568303
Jessica L Huang, Osman Sharifi, Dag H Yasui, Janine M LaSalle
{"title":"MeCP2 at the crossroads of hypoxia, oxidative stress, and gene regulation in Rett syndrome.","authors":"Jessica L Huang, Osman Sharifi, Dag H Yasui, Janine M LaSalle","doi":"10.1080/17501911.2025.2568303","DOIUrl":"https://doi.org/10.1080/17501911.2025.2568303","url":null,"abstract":"<p><p>Rett syndrome (RTT) is a severe neurodevelopmental disorder primarily affecting females, caused by mutations in the X-linked gene <i>MECP2</i>. This gene encodes methyl CpG binding protein 2 (MeCP2), a multifunctional epigenetic regulator critical for neuronal gene regulation. In addition to well-characterized neurological symptoms, such as seizures and motor abnormalities, RTT patients frequently present with irregular breathing patterns that induce intermittent hypoxia, suggesting that MeCP2 contributes to respiratory regulation as well as the brain's cellular and molecular response to hypoxia. Mechanistically, MeCP2 appears to influence hypoxia-induced expression of the neuroprotective peptide brain-derived neurotrophic factor (BDNF), as impaired BDNF regulation in MeCP2-deficient neurons contributes to hypoxia vulnerability. RTT patients also display increased oxidative stress, marked by elevated lipid peroxidation, DNA damage, and reduced antioxidant production. Dysfunctional mitochondria in MeCP2-deficient astrocytes and neurons further propagate oxidative damage and non-cell-autonomous effects of MeCP2 loss. Moreover, recent transcriptomic studies revealed widespread transcriptional dysregulation in RTT, including pathways associated with mitochondrial function and oxidative stress. We review and discuss an expanded role for MeCP2 as a critical integrator of hypoxia sensing, oxidative stress regulation, and transcriptional adaptation in the developing brain, offering new insights into treatments targeting the complex pathophysiology of RTT.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1-11"},"PeriodicalIF":2.6,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-01Epub Date: 2025-08-21DOI: 10.1080/17501911.2025.2548755
Sarah Williams, Susan J Clark, Ruth Pidsley, Clare Stirzaker
{"title":"DNA methylation biomarkers for the diagnosis and treatment management of breast cancer: where are we now?","authors":"Sarah Williams, Susan J Clark, Ruth Pidsley, Clare Stirzaker","doi":"10.1080/17501911.2025.2548755","DOIUrl":"10.1080/17501911.2025.2548755","url":null,"abstract":"<p><p>Breast cancer is one of the most commonly diagnosed cancers worldwide and is a significant contributor to the global cancer burden. It is a clinically heterogeneous disease and reliable tools are needed to support treatment decisions, including patient risk, prediction of therapeutic response and monitoring patients throughout their cancer journey. DNA methylation alterations are an early occurring, highly pervasive and stable modification during tumorigenesis, making DNA methylation an attractive target for the development of biomarkers. In this review, we first provide an overview of DNA methylation and explore its role in cancer, with an emphasis on breast cancer. We then focus on the potential use of tissue- and blood-based DNA methylation biomarkers to inform clinical decision-making in breast cancer paradigms: diagnosis; disease sub-typing; prediction of therapy response to neoadjuvant chemotherapy, endocrine therapy and immunotherapy; prognosis; and the tumor microenvironment. We highlight the significant progress achieved over recent decades in the development of DNA methylation-based biomarkers for breast cancer care. We end by discussing how the integration of advanced research methodologies and bioinformatic tools, and their incorporation into liquid biopsy platforms and ctDNA assays, offer promising opportunities for these biomarkers to be widely adopted in clinical practice.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1107-1122"},"PeriodicalIF":2.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144947520","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-01Epub Date: 2025-09-10DOI: 10.1080/17501911.2025.2558496
Chia-Hsuan Li, Bao-Yu Chen, Chih-Wei Lin, Shulan Hsieh, Cheng-Ta Yang, Joshua Oon Soo Goh, Yun-Hsuan Chang, Sheng-Hsiang Lin
{"title":"MicroRNAs signatures in small extracellular vesicles for psychological resilience in young adults using machine learning.","authors":"Chia-Hsuan Li, Bao-Yu Chen, Chih-Wei Lin, Shulan Hsieh, Cheng-Ta Yang, Joshua Oon Soo Goh, Yun-Hsuan Chang, Sheng-Hsiang Lin","doi":"10.1080/17501911.2025.2558496","DOIUrl":"10.1080/17501911.2025.2558496","url":null,"abstract":"<p><strong>Aims: </strong>Psychological resilience refers to an individual's capacity to adapt to adverse events. MicroRNAs (miRNAs) play a crucial role in regulating post-transcriptional processes, while small extracellular vesicles (sEVs) act as transport vehicles. This study aimed to employ genome-wide profiling to identify and validate differences in the expression of resilience-associated sEV-miRNAs between low resilience (LR) and high resilience (HR) in young adults.</p><p><strong>Methods: </strong>Eighty participants were divided into LR or HR based on the Connor - Davidson Resilience Scale (CD-RISC). The expression levels of the target sEV-miRNAs in LR and HR were compared and analyzed.</p><p><strong>Results: </strong>Expression analyses demonstrated significant differences in let-7b, miR-151b, miR-335, and miR-193a between LR and HR (<i>p</i> < 0.01), with let-7b showing the highest discriminative ability. The AUC values for each sEV-miRNA ranged from 0.74 to 0.94, based on logistic regression and three machine learning models: random forest, support vector machine, and eXtreme gradient boosting. Based on leave-one-out cross-validation in different models, the combined four sEV-miRNAs demonstrated strong performance for detecting LR (AUC = 0.87-0.90). Sex-specific differences were also observed, with female participants showing more pronounced resilience signatures in targeted sEV-miRNAs.</p><p><strong>Conclusions: </strong>These findings suggest that sEV-miRNAs hold potential as biomarkers for psychological resilience in young adults.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"1043-1055"},"PeriodicalIF":2.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145029180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EpigenomicsPub Date : 2025-10-01Epub Date: 2025-07-22DOI: 10.1080/17501911.2025.2535938
Md Sadique Hussain, Yumna Khan, Mudasir Maqbool, Gyas Khan, Wedad Mawkili, Faroq Kamli, Ali Hanbashi, Prawez Alam
{"title":"Epigenetic alterations in prostate cancer: the role of chromatin remodeling.","authors":"Md Sadique Hussain, Yumna Khan, Mudasir Maqbool, Gyas Khan, Wedad Mawkili, Faroq Kamli, Ali Hanbashi, Prawez Alam","doi":"10.1080/17501911.2025.2535938","DOIUrl":"10.1080/17501911.2025.2535938","url":null,"abstract":"<p><p>Prostate cancer (PCa) is one of the most common cancers in men, distinguished by a multifaceted pathogenesis that involves substantial epigenetic modifications. This article emphasizes the critical role of chromatin remodeling in PCa advancement. Chromatin remodeling, a key epigenetic mechanism, influences gene expression by modulating the chromatin structure through the action of specialized complexes such as SWI/SNF, ISWI, CHD, & INO80, thereby regulating genes vital to tumor progression. A comprehensive literature search was conducted using PubMed, Web of Science, and Scopus databases for studies published between 2000 and 2025, focusing on chromatin remodeling, epigenetic alterations, and therapeutic strategies in PCa. The review discusses the major epigenetic changes observed in PCa, including DNA and RNA methylation, histone modifications, and non-coding RNA (ncRNA)-mediated chromatin remodeling. The evolving epigenetic landscape shaped by these alterations offers insights into novel therapeutic opportunities. The clinical relevance of targeting chromatin remodeling complexes is explored, alongside existing therapies and potential future interventions. This review also addresses challenges in studying chromatin remodeling and highlights emerging technologies that could enhance understanding of PCa epigenetics. This comprehensive exploration underscores the promise of chromatin remodeling as both a biomarker and a therapeutic target in PCa management.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"967-991"},"PeriodicalIF":2.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12490391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144689660","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}