The Human Infertility Single-cell Testis Atlas (HISTA): An interactive molecular scRNA-Seq reference of the human testis

Background The Human Infertility Single-cell Testis Atlas (HISTA) is an interactive web tool and a reference for navigating the transcriptome of the human testis. It was developed using joint analyses of scRNA-Seq datasets derived from a dozen donors, including healthy adult controls, juveniles, and several infertility cases. HISTA is very different than other websites of testis scRNA-seq data, providing visualization and hypothesis testing tools on a batch-removed and integrated dataset of 23429 genes measured across 26093 cells using. Objective The main goal of this manuscript is to describe HISTA in detail and highlight its unique and novel features. Methods Therefore, we used HISTA as a guide for its application and demonstrated HISTA’s translational capacity to follow up on two observations of biological relevance. Results Our first analytical vignette identifies novel groupings of tightly regulated long non-coding RNA (lncRNA) molecules throughout spermatogenesis, suggesting specific functional genomics of these groupings. This analysis also found highly controlled expression of pairs of sense and antisense transcripts, suggesting conjoined regulatory mechanisms. In the next investigative vignette, we examined gene patterns in undifferentiated spermatogonia (USgs). We found the NANOS family of genes function as key drivers of transcriptomic signatures involved in human spermatogonial self-renewal programming; for the first time, demonstrating the relationship of NANOS1/2/3 transcripts in humans with scRNA-seq. Discussion and Conclusions Using HISTA, we found new observations that contribute to unraveling the mechanisms behind transcriptional regulation and maintenance germ cells across spermatogenesis. Furthermore, our findings provide guidance on future validation studies and experimental direction. Overall, HISTA continues to be utilized in testis-related research, and thus is updated regularly with new analytical methods, visualizations, and data. We aim to have it serve as a research environment for a broad range of investigators looking to explore the testis tissue and male infertility. Availability and Implementation HISTA is available as an interactive web tool: https://conradlab.shinyapps.io/HISTA Source code and documentation for HISTA are provided on GitHub: https://github.com/eisascience/HISTA

[1]  M. Surani,et al.  DMRT1 regulates human germline commitment , 2023, Nature Cell Biology.

[2]  Peng Yuan,et al.  DMRT1 drives the human germline forward , 2023, Nature Cell Biology.

[3]  Ashutosh Kumar Singh,et al.  Epigenetic Reprogramming in Mice and Humans: From Fertilization to Primordial Germ Cell Development , 2023, Cells.

[4]  P. Ping,et al.  Application of single-cell RNA sequencing on human testicular samples: a comprehensive review , 2023, International journal of biological sciences.

[5]  Jie Yu,et al.  Analysis of lncRNA and mRNA Expression Profiling in Immature and Mature DeZhou donkey (equine Taurus) Testes. , 2023, Reproduction in domestic animals = Zuchthygiene.

[6]  C. Gravholt,et al.  Testicular Architecture of Men with 46,XX Testicular Disorders of Sex Development , 2023, Sexual Development.

[7]  J. Yang,et al.  A single-cell transcriptomic landscape of mouse testicular aging , 2022, Journal of advanced research.

[8]  A. Kimura,et al.  Evidence for a functional role of Start, a long noncoding RNA, in mouse spermatocytes , 2022, PloS one.

[9]  M. J. Xavier,et al.  Diverse monogenic subforms of human spermatogenic failure , 2022, Nature Communications.

[10]  Yonatan B. Tzur lncRNAs in fertility: redefining the gene expression paradigm? , 2022, Trends in genetics : TIG.

[11]  Yabing Chen,et al.  LncRNA Tug1 maintains blood–testis barrier integrity by modulating Ccl2 expression in high-fat diet mice , 2022, Cellular and Molecular Life Sciences.

[12]  Huan Wang,et al.  Long Noncoding RNAs: Recent Insights into Their Role in Male Infertility and Their Potential as Biomarkers and Therapeutic Targets , 2021, International journal of molecular sciences.

[13]  D. Conrad,et al.  Comparative single-cell analysis of biopsies clarifies pathogenic mechanisms in Klinefelter syndrome. , 2021, American journal of human genetics.

[14]  K. Ohbo,et al.  Maintenance DNA methylation in pre-meiotic germ cells regulates meiotic prophase by facilitating homologous chromosome pairing. , 2021, Development.

[15]  S. Takada,et al.  A Testis-Specific Long Noncoding RNA, Start, Is a Regulator of Steroidogenesis in Mouse Leydig Cells , 2021, Frontiers in Endocrinology.

[16]  B. Cairns,et al.  Single-cell analysis of the developing human testis reveals somatic niche cell specification and fetal germline stem cell establishment. , 2021, Cell stem cell.

[17]  I. Santin,et al.  The Role of lncRNAs in Gene Expression Regulation through mRNA Stabilization , 2021, Non-coding RNA.

[18]  S. Rajender,et al.  Long non-coding RNAs (lncRNAs) in spermatogenesis and male infertility , 2020, Reproductive Biology and Endocrinology.

[19]  Evan Z. Macosko,et al.  Dissecting mammalian spermatogenesis using spatial transcriptomics , 2020, bioRxiv.

[20]  Raphael Gottardo,et al.  Integrated analysis of multimodal single-cell data , 2020, Cell.

[21]  P. Munusamy,et al.  Single-Cell RNA Sequencing of the Cynomolgus Macaque Testis Reveals Conserved Transcriptional Profiles during Mammalian Spermatogenesis. , 2020, Developmental cell.

[22]  Yanli Zhang,et al.  Expression pattern and potential role of Nanos3 in regulating testosterone biosynthesis in Leydig cells of sheep. , 2020, Theriogenology.

[23]  M. Sajek,et al.  Human NANOS1 Represses Apoptosis by Downregulating Pro-Apoptotic Genes in the Male Germ Cell Line , 2020, International journal of molecular sciences.

[24]  Dan Zhang,et al.  Construction of a human cell landscape at single-cell level , 2020, Nature.

[25]  Jun Z. Li,et al.  Single-cell RNA sequencing of human, macaque, and mouse testes uncovers conserved and divergent features of mammalian spermatogenesis , 2020, bioRxiv.

[26]  Jinming Huang,et al.  Long noncoding RNAs: new insights in modulating mammalian spermatogenesis , 2020, Journal of Animal Science and Biotechnology.

[27]  C. Lindskog,et al.  The Dynamic Transcriptional Cell Atlas of Testis Development during Human Puberty , 2020, Cell stem cell.

[28]  E. Levin,et al.  Cannabis use is associated with potentially heritable widespread changes in autism candidate gene DLGAP2 DNA methylation in sperm , 2019, Epigenetics.

[29]  Hong Chen,et al.  Analysis of Long Non-Coding RNA and mRNA Expression Profiling in Immature and Mature Bovine (Bos taurus) Testes , 2019, Front. Genet..

[30]  J. Marchini,et al.  Unified single-cell analysis of testis gene regulation and pathology in five mouse strains , 2019, eLife.

[31]  L. Laurent,et al.  The Neonatal and Adult Human Testis Defined at the Single-Cell Level , 2019, Cell reports.

[32]  Frédéric Chalmel,et al.  The ReproGenomics Viewer: a multi-omics and cross-species resource compatible with single-cell studies for the reproductive science community , 2019, Bioinform..

[33]  Oscar Franzén,et al.  PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data , 2019, Database J. Biol. Databases Curation.

[34]  Ellen K. Velte,et al.  The Mammalian Spermatogenesis Single-Cell Transcriptome, from Spermatogonial Stem Cells to Spermatids. , 2018, Cell reports.

[35]  Geoffrey J Maher,et al.  The adult human testis transcriptional cell atlas , 2018, Cell Research.

[36]  D. Monk,et al.  Epigenetic Symmetry of DLGAP2: Pre-Implantation Maternal Methylation Switches to a Random Monoallelic Profile in Somatic Tissues , 2018, OBM Genetics.

[37]  C. Cho,et al.  Profiling of testis-specific long noncoding RNAs in mice , 2018, BMC genomics.

[38]  L. Fañanás,et al.  Genetic variability in scaffolding proteins and risk for schizophrenia and autism-spectrum disorders: a systematic review. , 2018, Journal of psychiatry & neuroscience : JPN.

[39]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[40]  A. Kimura,et al.  A Testis-Specific Long Non-Coding RNA, lncRNA-Tcam1, Regulates Immune-Related Genes in Mouse Male Germ Cells , 2017, Front. Endocrinol..

[41]  D. D. de Rooij,et al.  Spermatogonial kinetics in humans , 2017, Development.

[42]  A. Silahtaroglu,et al.  The DLGAP family: neuronal expression, function and role in brain disorders , 2017, Molecular Brain.

[43]  E. Xu,et al.  LncRNA, a new component of expanding RNA-protein regulatory network important for animal sperm development. , 2016, Seminars in cell & developmental biology.

[44]  Jesse M. Engreitz,et al.  Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression , 2016, Nature Reviews Molecular Cell Biology.

[45]  Y. Saeys,et al.  SCORPIUS improves trajectory inference and identifies novel modules in dendritic cell development , 2016, bioRxiv.

[46]  M. McCarthy,et al.  Tensor decomposition for multi-tissue gene expression experiments , 2016, Nature Genetics.

[47]  F. Tang,et al.  The Transcriptome and DNA Methylome Landscapes of Human Primordial Germ Cells , 2015, Cell.

[48]  M. Albà,et al.  Long non-coding RNAs as a source of new peptides , 2014, eLife.

[49]  Michael Morse,et al.  Multiple knockout mouse models reveal lincRNAs are required for life and brain development , 2013, eLife.

[50]  J. Rinn,et al.  Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination , 2013, Nature Communications.

[51]  C. Wahlestedt,et al.  Regulation of chromatin structure by long noncoding RNAs: focus on natural antisense transcripts. , 2012, Trends in genetics : TIG.

[52]  Howard Y. Chang,et al.  Molecular mechanisms of long noncoding RNAs. , 2011, Molecular cell.

[53]  J. Rinn,et al.  Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. , 2011, Genes & development.

[54]  J. Clarke,et al.  What is a systematic review? , 2011, Evidence Based Nursing.

[55]  Y. Saga Function of Nanos2 in the male germ cell lineage in mice , 2010, Cellular and Molecular Life Sciences.

[56]  C. Wahlestedt,et al.  Regulatory roles of natural antisense transcripts , 2009, Nature Reviews Molecular Cell Biology.

[57]  E. Birney,et al.  Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt , 2009, Nature Protocols.

[58]  A. Latos-Bieleńska,et al.  The highly conserved NANOS2 protein: testis-specific expression and significance for the human male reproduction. , 2009, Molecular human reproduction.

[59]  P. Houillier,et al.  A role for Rhesus factor Rhcg in renal ammonium excretion and male fertility , 2008, Nature.

[60]  R. Geremia,et al.  Potential role of Nanos3 in maintaining the undifferentiated spermatogonia population. , 2008, Developmental biology.

[61]  A. Hartemink,et al.  Computational and experimental identification of novel human imprinted genes. , 2007, Genome research.

[62]  Y. Saga,et al.  Functional redundancy among Nanos proteins and a distinct role of Nanos2 during male germ cell development , 2007, Development.

[63]  Haifan Lin,et al.  Nanos Maintains Germline Stem Cell Self-Renewal by Preventing Differentiation , 2004, Science.

[64]  K. Abe,et al.  Conserved Role of nanos Proteins in Germ Cell Development , 2003, Science.

[65]  Y. Saga,et al.  nanos1: a mouse nanos gene expressed in the central nervous system is dispensable for normal development , 2003, Mechanisms of Development.

[66]  T. Omi,et al.  A Japanese propositus with D-- phenotype characterized by the deletion of both the RHCE gene and D1S80 locus situated in chromosome 1p and the existence of a new CE-D-CE hybrid gene , 2000, Journal of Human Genetics.

[67]  Giles R. Scuderi,et al.  A Comprehensive Review , 2017 .

[68]  K. Morris Long Non-coding RNAs in Human Disease , 2016, Current Topics in Microbiology and Immunology.

[69]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[70]  Cedric E. Ginestet ggplot2: Elegant Graphics for Data Analysis , 2011 .