Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 161(6): 1437-52, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046443

ABSTRACT

Germ cells are vital for transmitting genetic information from one generation to the next and for maintaining the continuation of species. Here, we analyze the transcriptome of human primordial germ cells (PGCs) from the migrating stage to the gonadal stage at single-cell and single-base resolutions. Human PGCs show unique transcription patterns involving the simultaneous expression of both pluripotency genes and germline-specific genes, with a subset of them displaying developmental-stage-specific features. Furthermore, we analyze the DNA methylome of human PGCs and find global demethylation of their genomes. Approximately 10 to 11 weeks after gestation, the PGCs are nearly devoid of any DNA methylation, with only 7.8% and 6.0% of the median methylation levels in male and female PGCs, respectively. Our work paves the way toward deciphering the complex epigenetic reprogramming of the germline with the aim of restoring totipotency in fertilized oocytes.


Subject(s)
DNA Methylation , Germ Cells/metabolism , Transcriptome , Cell Movement , Chromosomes, Human, X , Cluster Analysis , Embryo, Mammalian/metabolism , Female , Histones/metabolism , Humans , Male , Principal Component Analysis , SOX Transcription Factors/metabolism
2.
Nucleic Acids Res ; 50(13): 7479-7492, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35819189

ABSTRACT

Genome assembly has been benefited from long-read sequencing technologies with higher accuracy and higher continuity. However, most human genome assembly require large amount of DNAs from homogeneous cell lines without keeping cell heterogeneities, since cell heterogeneity could profoundly affect haplotype assembly results. Herein, using single-cell genome long-read sequencing technology (SMOOTH-seq), we have sequenced K562 and HG002 cells on PacBio HiFi and Oxford Nanopore Technologies (ONT) platforms and conducted de novo genome assembly. For the first time, we have completed the human genome assembly with high continuity (with NG50 of ∼2 Mb using 95 individual K562 cells) at single-cell levels, and explored the impact of different assemblers and sequencing strategies on genome assembly. With sequencing data from 30 diploid individual HG002 cells of relatively high genome coverage (average coverage ∼41.7%) on ONT platform, the NG50 can reach over 1.3 Mb. Furthermore, with the assembled genome from K562 single-cell dataset, more complete and accurate set of insertion events and complex structural variations could be identified. This study opened a new chapter on the practice of single-cell genome de novo assembly.


Subject(s)
Genome, Human , Nanopores , Chromosome Mapping , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods
3.
FASEB J ; 36(2): e22131, 2022 02.
Article in English | MEDLINE | ID: mdl-34985827

ABSTRACT

Although germ cell fate is believed to be determined by signaling factors from differentiated somatic cells, the molecular mechanism behind this process remains obscure. In this study, premature meiosis in male germ cells was observed during the embryonic stage by conditional activation of ß-catenin in Sertoli cells. Somatic and germ cell transcriptome results indicated that the BMP signaling pathway was enriched after ß-catenin activation. In addition, we observed a decreased DNA methylation within a reduction of DNMT3A in germ cells of ß-catenin activated testes and reversed increase after inhibiting BMP signaling pathway with LDN-193189. We also found that Dazl expression was increased in ß-catenin activated testes and decreased after LDN treatment. Taken together, this study demonstrates that male germ cells entered meiosis prematurely during the embryonic stage after ß-catenin activated in Sertoli cells. BMP signaling pathway involved in germ cell meiosis initiation by mediating DNA methylation to induce meiotic genes expression.


Subject(s)
Bone Morphogenetic Proteins/genetics , Embryonic Development/genetics , Germ Cells/physiology , Meiosis/genetics , RNA-Binding Proteins/genetics , Up-Regulation/genetics , Animals , Cell Differentiation/genetics , DNA Methylation/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Sertoli Cells/physiology , Signal Transduction/genetics , Testis/pathology , Transcriptome/genetics , beta Catenin/genetics
4.
PLoS Biol ; 17(7): e3000365, 2019 07.
Article in English | MEDLINE | ID: mdl-31269016

ABSTRACT

The developmental pathway of the neural retina (NR) and retinal pigment epithelium (RPE) has been revealed by extensive research in mice. However, the molecular mechanisms underlying the development of the human NR and RPE, as well as the interactions between these two tissues, have not been well defined. Here, we analyzed 2,421 individual cells from human fetal NR and RPE using single-cell RNA sequencing (RNA-seq) technique and revealed the tightly regulated spatiotemporal gene expression network of human retinal cells. We identified major cell classes of human fetal retina and potential crucial transcription factors for each cell class. We dissected the dynamic expression patterns of visual cycle- and ligand-receptor interaction-related genes in the RPE and NR. Moreover, we provided a map of disease-related genes for human fetal retinal cells and highlighted the importance of retinal progenitor cells as potential targets of inherited retinal diseases. Our findings captured the key in vivo features of the development of the human NR and RPE and offered insightful clues for further functional studies.


Subject(s)
Gene Expression Regulation, Developmental , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome , Adult , Cell Cycle/genetics , Cells, Cultured , Cluster Analysis , Gene Expression Profiling/methods , Gene Ontology , Humans , Retina/cytology , Retina/embryology , Retinal Diseases/genetics , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/embryology
5.
PLoS Biol ; 17(4): e3000201, 2019 04.
Article in English | MEDLINE | ID: mdl-30933975

ABSTRACT

Cellular senescence is a driver of various aging-associated disorders, including osteoarthritis. Here, we identified a critical role for Yes-associated protein (YAP), a major effector of Hippo signaling, in maintaining a younger state of human mesenchymal stem cells (hMSCs) and ameliorating osteoarthritis in mice. Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated protein 9 nuclease (Cas9)-mediated knockout (KO) of YAP in hMSCs resulted in premature cellular senescence. Mechanistically, YAP cooperated with TEA domain transcriptional factor (TEAD) to activate the expression of forkhead box D1 (FOXD1), a geroprotective protein. YAP deficiency led to the down-regulation of FOXD1. In turn, overexpression of YAP or FOXD1 rejuvenated aged hMSCs. Moreover, intra-articular administration of lentiviral vector encoding YAP or FOXD1 attenuated the development of osteoarthritis in mice. Collectively, our findings reveal YAP-FOXD1, a novel aging-associated regulatory axis, as a potential target for gene therapy to alleviate osteoarthritis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Forkhead Transcription Factors/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Proliferation/genetics , Cellular Senescence/physiology , Forkhead Transcription Factors/genetics , Heterografts , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Osteoarthritis/genetics , Signal Transduction , Transcription Factors/genetics , Transcriptional Activation , Up-Regulation , YAP-Signaling Proteins
6.
BMC Genomics ; 21(1): 438, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32590948

ABSTRACT

BACKGROUND: Sex-related genes play a crucial role in gonadal differentiation into testes or ovaries. However, the genetic control of gonadal differentiation in Muscovy ducks remains unknown. Therefore, the objective of our study was to screen new candidate genes associated with ovarian and testicular development. RESULTS: In this study, 24 males before gonadal differentiation (MB), 24 females before gonadal differentiation (FB), 24 males after gonadal differentiation (MA) and 24 females after gonadal differentiation (FA) were selected from Putian Muscovy ducks, forming 4 groups. RNA-Seq revealed 101.76 Gb of clean reads and 2800 differentially expressed genes (DEGs), including 46 in MB vs FB, 609 in MA vs FA, 1027 in FA vs FB, and 1118 in MA vs MB. A total of 146 signalling pathways were enriched by KEGG analysis, among which 20, 108, 108 and 116 signalling pathways were obtained in MB vs FB, MA vs MB, MA vs FA and FA vs FB, respectively. In further GO and KEGG analyses, a total of 21 candidate genes related to gonad differentiation and development in Muscovy ducks were screened. Among these, 9 genes were involved in the differentiation and development of the testes, and 12 genes were involved in the differentiation and development of the ovaries. In addition, RNA-Seq data revealed 2744 novel genes. CONCLUSIONS: RNA-Seq data revealed 21 genes related to gonadal differentiation and development in Muscovy ducks. We further identified 12 genes, namely, WNT5B, HTRA3, RSPO3, BMP3, HNRNPK, NIPBL, CREB3L4, DKK3, UBE2R2, UBPL3KCMF1, ANXA2, and OSR1, involved in the differentiation and development of ovaries. Moreover, 9 genes, namely, TTN, ATP5A1, DMRT1, DMRT3, AMH, MAP3K1, PIK3R1, AGT and ADAMTSL1, were related to the differentiation and development of testes. Moreover, after gonadal differentiation, DMRT3, AMH, PIK3R1, ADAMTSL1, AGT and TTN were specifically highly expressed in males. WNT5B, ANXA2 and OSR1 were specifically highly expressed in females. These results provide valuable information for studies on the sex control of Muscovy ducks and reveal novel candidate genes for the differentiation and development of testes and ovaries.


Subject(s)
Ducks/growth & development , Gene Expression Profiling/veterinary , Gonads/physiology , Animals , Cell Differentiation , Ducks/genetics , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Gonads/growth & development , Male , Sequence Analysis, RNA , Sex Characteristics
7.
Ann Rheum Dis ; 78(1): 100-110, 2019 01.
Article in English | MEDLINE | ID: mdl-30026257

ABSTRACT

OBJECTIVES: Understanding the molecular mechanisms underlying human cartilage degeneration and regeneration is helpful for improving therapeutic strategies for treating osteoarthritis (OA). Here, we report the molecular programmes and lineage progression patterns controlling human OA pathogenesis using single-cell RNA sequencing (scRNA-seq). METHODS: We performed unbiased transcriptome-wide scRNA-seq analysis, computational analysis and histological assays on 1464 chondrocytes from 10 patients with OA undergoing knee arthroplasty surgery. We investigated the relationship between transcriptional programmes of the OA landscape and clinical outcome using severity index and correspondence analysis. RESULTS: We identified seven molecularly defined populations of chondrocytes in the human OA cartilage, including three novel phenotypes with distinct functions. We presented gene expression profiles at different OA stages at single-cell resolution. We found a potential transition among proliferative chondrocytes, prehypertrophic chondrocytes and hypertrophic chondrocytes (HTCs) and defined a new subdivision within HTCs. We revealed novel markers for cartilage progenitor cells (CPCs) and demonstrated a relationship between CPCs and fibrocartilage chondrocytes using computational analysis. Notably, we derived predictive targets with respect to clinical outcomes and clarified the role of different cell types for the early diagnosis and treatment of OA. CONCLUSIONS: Our results provide new insights into chondrocyte taxonomy and present potential clues for effective and functional manipulation of human OA cartilage regeneration that could lead to improved health.


Subject(s)
Chondrocytes/metabolism , Osteoarthritis, Knee/genetics , Sequence Analysis, RNA , Cartilage, Articular/cytology , Chondrogenesis/genetics , Computational Biology , Disease Progression , Gene Expression Profiling , Humans , Phenotype , Stem Cells/metabolism , Transcriptome
8.
J Biol Chem ; 292(43): 17577-17586, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28900034

ABSTRACT

Sertoli and granulosa cells are two major types of somatic cells in male and female gonads, respectively. Previous studies have shown that Sertoli and granulosa cells are derived from common progenitor cells and that differentiation of these two cell types is regulated by sex differentiation genes. The signaling pathway including the adhesion and transcription factor Ctnnb1 (cadherin-associated protein, ß1, also known as ß-catenin) regulates differentiation of granulosa cells in the absence of the transcription factor Sry, and overactivation of ß-catenin in the presence of Sry leads to granulosa prior to sex determination. Surprisingly, our previous study found that ß-catenin overactivation in Sertoli cells after sex determination can also cause disruption of the testicular cord and aberrant testis development. However, the underlying molecular mechanism was unclear. In this study, we found that constitutive activation of Ctnnb1 in Sertoli cells led to ectopic expression of the granulosa cell-specific marker FOXL2 in testes. Co-staining experiments revealed that FOXL2-positive cells were derived from Sertoli cells, and Sertoli cells were transformed into granulosa-like cells after Ctnnb1 overactivation. Further studies demonstrated that CTNNB1 induced Foxl2 expression by directly binding to transcription factor Tcf/Lef-binding sites in the FOXL2 promoter region. We also found that direct overexpression of Foxl2 decreased the expression of Sertoli cell-specific genes in primary Sertoli cells. Taken together, these results demonstrate that repression of ß-catenin (CTNNB1) signaling is required for lineage maintenance of Sertoli cells. Our study provides a new mechanism for Sertoli cell lineage maintenance during gonad development.


Subject(s)
Cell Transdifferentiation , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation , Granulosa Cells/metabolism , Sertoli Cells/metabolism , Signal Transduction , beta Catenin/biosynthesis , Animals , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Granulosa Cells/cytology , Male , Mice , Mice, Transgenic , Sertoli Cells/cytology , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , beta Catenin/genetics
9.
Cancer Discov ; 14(6): 1082-1105, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38445965

ABSTRACT

Colorectal cancer is a highly heterogeneous disease, with well-characterized subtypes based on genome, DNA methylome, and transcriptome signatures. To chart the epigenetic landscape of colorectal cancers, we generated a high-quality single-cell chromatin accessibility atlas of epithelial cells for 29 patients. Abnormal chromatin states acquired in adenomas were largely retained in colorectal cancers, which were tightly accompanied by opposite changes of DNA methylation. Unsupervised analysis on malignant cells revealed two epigenetic subtypes, exactly matching the iCMS classification, and key iCMS-specific transcription factors (TFs) were identified, including HNF4A and PPARA for iCMS2 tumors and FOXA3 and MAFK for iCMS3 tumors. Notably, subtype-specific TFs bind to distinct target gene sets and contribute to both interpatient similarities and diversities for both chromatin accessibilities and RNA expressions. Moreover, we identified CpG-island methylator phenotypes and pinpointed chromatin state signatures and TF regulators for the CIMP-high subtype. Our work systematically revealed the epigenetic basis of the well-known iCMS and CIMP classifications of colorectal cancers. SIGNIFICANCE: Our work revealed the epigenetic basis of the well-known iCMS and CIMP classifications of colorectal cancers. Moreover, interpatient minor similarities and major diversities of chromatin accessibility signatures of TF target genes can faithfully explain the corresponding interpatient minor similarities and major diversities of RNA expression signatures of colorectal cancers, respectively. This article is featured in Selected Articles from This Issue, p. 897.


Subject(s)
Chromatin , Colorectal Neoplasms , Epigenesis, Genetic , Single-Cell Analysis , Transcription Factors , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Chromatin/genetics , Chromatin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic
10.
Nat Cell Biol ; 26(9): 1458-1469, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39080410

ABSTRACT

The conversion of DNA 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by TET enzymes represents a significant epigenetic modification, yet its role in early human embryos remains largely unknown. Here we showed that the early human embryo inherited a significant amount of 5hmCs from an oocyte, which unexpectedly underwent de novo hydroxymethylation during its growth. Furthermore, the generation of 5hmC in the paternal genome after fertilization roughly followed the maternal pattern, which was linked to DNA methylation dynamics and regions of sustained methylation. The 5hmCs persisted until the eight-cell stage and exhibited high enrichment at OTX2 binding sites, whereas knockdown of OTX2 in human embryos compromised the expression of early lineage genes. Specifically, the depletion of 5hmC affected the activation of embryonic genes, which was further evaluated by ectopically expressing mouse Tet3 in human early embryos. These findings revealed distinct dynamics of 5hmC and unravelled its multifaceted functions in early human embryonic development.


Subject(s)
5-Methylcytosine , Cytosine , DNA Methylation , DNA-Binding Proteins , Dioxygenases , Embryonic Development , Gene Expression Regulation, Developmental , Otx Transcription Factors , Proto-Oncogene Proteins , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Humans , Animals , Otx Transcription Factors/metabolism , Otx Transcription Factors/genetics , Mice , Dioxygenases/metabolism , Dioxygenases/genetics , Cytosine/analogs & derivatives , Cytosine/metabolism , Embryonic Development/genetics , Female , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Male , Blastocyst/metabolism , Cell Lineage/genetics , Oocytes/metabolism , Epigenesis, Genetic , Binding Sites
11.
Cell Res ; 32(4): 333-348, 2022 04.
Article in English | MEDLINE | ID: mdl-35079138

ABSTRACT

Arteriogenesis rather than unspecialized capillary expansion is critical for restoring effective circulation to compromised tissues in patients. Deciphering the origin and specification of arterial endothelial cells during embryonic development will shed light on the understanding of adult arteriogenesis. However, during early embryonic angiogenesis, the process of endothelial diversification and molecular events underlying arteriovenous fate settling remain largely unresolved in mammals. Here, we constructed the single-cell transcriptomic landscape of vascular endothelial cells (VECs) during the time window for the occurrence of key vasculogenic and angiogenic events in both mouse and human embryos. We uncovered two distinct arterial VEC types, the major artery VECs and arterial plexus VECs, and unexpectedly divergent arteriovenous characteristics among VECs that are located in morphologically undistinguishable vascular plexus intra-embryonically. Using computational prediction and further lineage tracing of venous-featured VECs with a newly developed Nr2f2CrexER mouse model and a dual recombinase-mediated intersectional genetic approach, we revealed early and widespread arterialization from the capillaries with considerable venous characteristics. Altogether, our findings provide unprecedented and comprehensive details of endothelial heterogeneity and lineage relationships at early angiogenesis stages, and establish a new model regarding the arteriogenesis behaviors of early intra-embryonic vasculatures.


Subject(s)
Endothelial Cells , Neovascularization, Pathologic , Animals , Cell Differentiation , Humans , Mammals , Mice
12.
Medicine (Baltimore) ; 100(8): e24528, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33663059

ABSTRACT

BACKGROUND: Functional dyspepsia (FD) is a common and frequently-occurring disease in internal medicine. It is known that Liujunzi decoction and acupuncture are widely used in the treatment of FD, but there are few studies on the combination of Liujunzi decoction and acupuncture in the treatment of FD, and its safety and efficacy are still controversial. Therefore, the purpose of this study is to evaluate the efficacy and safety of acupuncture combined with Liujunzi decoction in the treatment of FD. METHODS: We designed a prospective randomized controlled trial. The study protocol was approved by the Clinical Research Ethics Committee of our hospital. Patients with FD were randomly assigned to the treatment group of acupuncture combined with Liujunzi Decoction (the experimental group) and the treatment group of Liujunzi Decoction (the control group) in a ratio of 1:1. Outcome indicators were Nepean Dyspepsia Index, the MOS item short from health survey, and adverse reactions. Finally, SPSS 18.0 software would be used for statistical analysis of the data. DISCUSSION: This study will evaluate the efficacy and safety of acupuncture combined with Liujunzi Decoction in the treatment of FD and provide clinical basis for the use of acupuncture combined with Liujunzi Decoction in the treatment of FD. OSF REGISTRATION NUMBER: DOI 10.17605/OSF.IO/67GKN.


Subject(s)
Acupuncture Therapy/methods , Drugs, Chinese Herbal/therapeutic use , Dyspepsia/therapy , Adolescent , Adult , Aged , Combined Modality Therapy , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/adverse effects , Dyspepsia/drug therapy , Humans , Middle Aged , Prospective Studies , Quality of Life , Research Design , Young Adult
13.
Nat Commun ; 12(1): 6839, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824237

ABSTRACT

Mammalian male germ cell development is a stepwise cell-fate transition process; however, the full-term developmental profile of male germ cells remains undefined. Here, by interrogating the high-precision transcriptome atlas of 11,598 cells covering 28 critical time-points, we demonstrate that cell-fate transition from mitotic to post-mitotic primordial germ cells is accompanied by transcriptome-scale reconfiguration and a transitional cell state. Notch signaling pathway is essential for initiating mitotic arrest and the maintenance of male germ cells' identities. Ablation of HELQ induces developmental arrest and abnormal transcriptome reprogramming of male germ cells, indicating the importance of cell cycle regulation for proper cell-fate transition. Finally, systematic human-mouse comparison reveals potential regulators whose deficiency contributed to human male infertility via mitotic arrest regulation. Collectively, our study provides an accurate and comprehensive transcriptome atlas of the male germline cycle and allows for an in-depth understanding of the cell-fate transition and determination underlying male germ cell development.


Subject(s)
Spermatozoa/cytology , Spermatozoa/growth & development , Animals , Gene Expression Regulation, Developmental , Humans , Male , Meiosis/genetics , Mice , Mitosis/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Spermatogenesis/genetics , Spermatozoa/metabolism , Transcriptome
14.
Cell Res ; 30(5): 376-392, 2020 05.
Article in English | MEDLINE | ID: mdl-32203131

ABSTRACT

Hematopoietic stem cells (HSCs) in adults are believed to be born from hemogenic endothelial cells (HECs) in mid-gestational embryos. Due to the rare and transient nature, the HSC-competent HECs have never been stringently identified and accurately captured, let alone their genuine vascular precursors. Here, we first used high-precision single-cell transcriptomics to unbiasedly examine the relevant EC populations at continuous developmental stages with intervals of 0.5 days from embryonic day (E) 9.5 to E11.0. As a consequence, we transcriptomically identified two molecularly different arterial EC populations and putative HSC-primed HECs, whose number peaked at E10.0 and sharply decreased thereafter, in the dorsal aorta of the aorta-gonad-mesonephros (AGM) region. Combining computational prediction and in vivo functional validation, we precisely captured HSC-competent HECs by the newly constructed Neurl3-EGFP reporter mouse model, and realized the enrichment further by a combination of surface markers (Procr+Kit+CD44+, PK44). Surprisingly, the endothelial-hematopoietic dual potential was rarely but reliably witnessed in the cultures of single HECs. Noteworthy, primitive vascular ECs from E8.0 experienced two-step fate choices to become HSC-primed HECs, namely an initial arterial fate choice followed by a hemogenic fate conversion. This finding resolves several previously observed contradictions. Taken together, comprehensive understanding of endothelial evolutions and molecular programs underlying HSC-primed HEC specification in vivo will facilitate future investigations directing HSC production in vitro.


Subject(s)
Aorta/embryology , Hemangioblasts/cytology , Hematopoiesis , Transcriptome , Animals , Cells, Cultured , Embryo, Mammalian , Mice , Mice, Inbred C57BL , Primary Cell Culture , Single-Cell Analysis
15.
Natl Sci Rev ; 7(8): 1306-1318, 2020 Aug.
Article in English | MEDLINE | ID: mdl-34692159

ABSTRACT

Brain tumors are among the most challenging human tumors for which the mechanisms driving progression and heterogeneity remain poorly understood. We combined single-cell RNA-seq with multi-sector biopsies to sample and analyze single-cell expression profiles of gliomas from 13 Chinese patients. After classifying individual cells, we generated a spatial and temporal landscape of glioma that revealed the patterns of invasion between the different sub-regions of gliomas. We also used single-cell inferred copy number variations and pseudotime trajectories to inform on the crucial branches that dominate tumor progression. The dynamic cell components of the multi-region biopsy analysis allowed us to spatially deconvolute with unprecedented accuracy the transcriptomic features of the core and those of the periphery of glioma at single-cell level. Through this rich and geographically detailed dataset, we were also able to characterize and construct the chemokine and chemokine receptor interactions that exist among different tumor and non-tumor cells. This study provides the first spatial-level analysis of the cellular states that characterize human gliomas. It also presents an initial molecular map of the cross-talks between glioma cells and the surrounding microenvironment with single-cell resolution.

16.
Sex Dev ; 13(3): 137-142, 2019.
Article in English | MEDLINE | ID: mdl-31450230

ABSTRACT

Under the same feeding conditions, the growth and development of male Muscovy ducks is significantly greater than that of females. Thus, controlling their sex expression can have economic benefits. However, reports on the degree of sex reversal in Muscovy ducks are scarce. In this study, we obtained sex-reversed Muscovy ducks by injecting letrozole before sex differentiation. We analyzed the degree of sex reversal in Muscovy ducks in terms of hormone levels, gonadal tissue development, and growth and found that the estradiol levels of AI-females (letrozole-induced female-to-male sex reversal) were not significantly different from those of normal males (p > 0.05), but the testosterone levels were significantly lower than those in normal males (p < 0.05). AI-female gonad tissue had changed, and the right gonad presented ovotestis tissue. The growth and development of AI-females was significantly less than that of normal males (p < 0.05) but was not significantly different from that of normal females (p > 0.05). Letrozole can induce female Muscovy ducks to convert into males, but the reversal cannot be completed. Thus, further studies are needed to elucidate how to entirely attain the change.


Subject(s)
Aromatase Inhibitors/adverse effects , Disorders of Sex Development/chemically induced , Ducks/physiology , Animals , Body Weight , Disorders of Sex Development/blood , Disorders of Sex Development/pathology , Ducks/growth & development , Female , Gonadal Steroid Hormones/blood , Gonads/pathology , Male
17.
Cell Rep ; 26(7): 1934-1950.e5, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759401

ABSTRACT

The heart is the central organ of the circulatory system, and its proper development is vital for maintaining human life. Here, we used single-cell RNA sequencing to profile the gene expression landscapes of ∼4,000 cardiac cells from human embryos and identified four major types of cells: cardiomyocytes (CMs), cardiac fibroblasts, endothelial cells (ECs), and valvar interstitial cells (VICs). Atrial and ventricular CMs acquired distinct features early in heart development. Furthermore, both CMs and fibroblasts show stepwise changes in gene expression. As development proceeds, VICs may be involved in the remodeling phase, and ECs display location-specific characteristics. Finally, we compared gene expression profiles between humans and mice and identified a series of unique features of human heart development. Our study lays the groundwork for elucidating the mechanisms of in vivo human cardiac development and provides potential clues to understand cardiac regeneration.


Subject(s)
Heart/physiopathology , Transcriptome/genetics , Female , Fetus , Humans , Male , Pregnancy
18.
Genome Biol ; 19(1): 31, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540203

ABSTRACT

BACKGROUND: Organogenesis is crucial for proper organ formation during mammalian embryonic development. However, the similarities and shared features between different organs and the cellular heterogeneity during this process at single-cell resolution remain elusive. RESULTS: We perform single-cell RNA sequencing analysis of 1916 individual cells from eight organs and tissues of E9.5 to E11.5 mouse embryos, namely, the forebrain, hindbrain, skin, heart, somite, lung, liver, and intestine. Based on the regulatory activities rather than the expression patterns, all cells analyzed can be well classified into four major groups with epithelial, mesodermal, hematopoietic, and neuronal identities. For different organs within the same group, the similarities and differences of their features and developmental paths are revealed and reconstructed. CONCLUSIONS: We identify mutual interactions between epithelial and mesenchymal cells and detect epithelial cells with prevalent mesenchymal features during organogenesis, which are similar to the features of intermediate epithelial/mesenchymal cells during tumorigenesis. The comprehensive transcriptome at single-cell resolution profiled in our study paves the way for future mechanistic studies of the gene-regulatory networks governing mammalian organogenesis.


Subject(s)
Epithelium/metabolism , Mesoderm/metabolism , Organogenesis/genetics , Transcriptome , Animals , Epithelial Cells/metabolism , Epithelium/embryology , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Mesoderm/embryology , Mice , Neurons/metabolism , Sequence Analysis, RNA , Single-Cell Analysis
20.
Cell Stem Cell ; 23(4): 599-614.e4, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30174296

ABSTRACT

Spermatogenesis generates mature male gametes and is critical for the proper transmission of genetic information between generations. However, the developmental landscapes of human spermatogenesis remain unknown. Here, we performed single-cell RNA sequencing (scRNA-seq) analysis for 2,854 testicular cells from donors with normal spermatogenesis and 174 testicular cells from one nonobstructive azoospermia (NOA) donor. A hierarchical model was established, which was characterized by the sequential and stepwise development of three spermatogonia subtypes, seven spermatocyte subtypes, and four spermatid subtypes. Further analysis identified several stage-specific marker genes of human germ cells, such as HMGA1, PIWIL4, TEX29, SCML1, and CCDC112. Moreover, we identified altered gene expression patterns in the testicular somatic cells of one NOA patient via scRNA-seq analysis, paving the way for further diagnosis of male infertility. Our work allows for the reconstruction of transcriptional programs inherent to sequential cell fate transition during human spermatogenesis and has implications for deciphering male-related reproductive disorders.


Subject(s)
Cell Lineage , Sequence Analysis, RNA , Single-Cell Analysis , Spermatogenesis/genetics , Spermatozoa/cytology , Spermatozoa/metabolism , Cells, Cultured , Computational Biology , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL