Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Sci China Life Sci ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39037697

ABSTRACT

The TET family is well known for active DNA demethylation and plays important roles in regulating transcription, the epigenome and development. Nevertheless, previous studies using knockdown (KD) or knockout (KO) models to investigate the function of TET have faced challenges in distinguishing its enzymatic and nonenzymatic roles, as well as compensatory effects among TET family members, which has made the understanding of the enzymatic role of TET not accurate enough. To solve this problem, we successfully generated mice catalytically inactive for specific Tet members (Tetm/m). We observed that, compared with the reported KO mice, mutant mice exhibited distinct developmental defects, including growth retardation, sex imbalance, infertility, and perinatal lethality. Notably, Tetm/m mouse embryonic stem cells (mESCs) were successfully established but entered an impaired developmental program, demonstrating extended pluripotency and defects in ectodermal differentiation caused by abnormal DNA methylation. Intriguingly, Tet3, traditionally considered less critical for mESCs due to its lower expression level, had a significant impact on the global hydroxymethylation, gene expression, and differentiation potential of mESCs. Notably, there were common regulatory regions between Tet1 and Tet3 in pluripotency regulation. In summary, our study provides a more accurate reference for the functional mechanism of Tet hydroxymethylase activity in mouse development and ESC pluripotency regulation.

2.
Article in English | MEDLINE | ID: mdl-38955498

ABSTRACT

The development and maturation of follicles is a sophisticated and multistage process. The dynamic gene expression of oocytes and their surrounding somatic cells and the dialogs between these cells are critical to this process. In this study, we accurately classified the oocyte and follicle development into nine stages and profiled the gene expression of mouse oocytes and their surrounding granulosa cells and cumulus cells. The clustering of the transcriptomes showed the trajectories of two distinct development courses of oocytes and their surrounding somatic cells. Gene expression changes precipitously increased at Type 4 stage and drastically dropped afterward within both oocytes and granulosa cells. Moreover, the number of differentially expressed genes between oocytes and granulosa cells dramatically increased at Type 4 stage, most of which persistently passed on to the later stages. Strikingly, cell communications within and between oocytes and granulosa cells became active from Type 4 stage onward. Cell dialogs connected oocytes and granulosa cells in both unidirectional and bidirectional manners. TGFB2/3, TGFBR2/3, INHBA/B, and ACVR1/1B/2B of TGF-ß signaling pathway functioned in the follicle development. NOTCH signaling pathway regulated the development of granulosa cells. Additionally, many maternally DNA methylation- or H3K27me3-imprinted genes remained active in granulosa cells but silent in oocytes during oogenesis. Collectively, Type 4 stage is the key turning point when significant transcription changes diverge the fate of oocytes and granulosa cells, and the cell dialogs become active to assure follicle development. These findings shed new insights on the transcriptome dynamics and cell dialogs facilitating the development and maturation of oocytes and follicles.


Subject(s)
Granulosa Cells , Oocytes , Ovarian Follicle , Transcriptome , Animals , Female , Oocytes/metabolism , Oocytes/growth & development , Oocytes/cytology , Mice , Granulosa Cells/metabolism , Granulosa Cells/cytology , Transcriptome/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/cytology , Cell Communication/genetics , Signal Transduction/genetics , Gene Expression Profiling/methods , DNA Methylation/genetics , Oogenesis/genetics
4.
Dev Cell ; 59(9): 1146-1158.e6, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38574734

ABSTRACT

Transcription factors (TFs) play important roles in early embryonic development, but factors regulating TF action, relationships in signaling cascade, genome-wide localizations, and impacts on cell fate transitions during this process have not been clearly elucidated. In this study, we used uliCUT&RUN-seq to delineate a TFAP2C-centered regulatory network, showing that it involves promoter-enhancer interactions and regulates TEAD4 and KLF5 function to mediate cell polarization. Notably, we found that maternal retinoic acid metabolism regulates TFAP2C expression and function by inducing the active demethylation of SINEs, indicating that the RARG-TFAP2C-TEAD4/KLF5 axis connects the maternal-to-zygotic transition to polarization. Moreover, we found that both genomic imprinting and SNP-transferred genetic information can influence TF positioning to regulate parental gene expressions in a sophisticated manner. In summary, we propose a ternary model of TF regulation in murine embryonic development with TFAP2C as the core element and metabolic, epigenetic, and genetic information as nodes connecting the pathways.


Subject(s)
Embryo Implantation , Gene Expression Regulation, Developmental , Transcription Factor AP-2 , Transcription Factors , Animals , Female , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Embryo Implantation/genetics , Embryonic Development/genetics , Gene Regulatory Networks , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Promoter Regions, Genetic/genetics , TEA Domain Transcription Factors/metabolism , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Tretinoin/metabolism
5.
Cell Rep ; 43(5): 114136, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38643480

ABSTRACT

Embryos, originating from fertilized eggs, undergo continuous cell division and differentiation, accompanied by dramatic changes in transcription, translation, and metabolism. Chromatin regulators, including transcription factors (TFs), play indispensable roles in regulating these processes. Recently, the trophoblast regulator TFAP2C was identified as crucial in initiating early cell fate decisions. However, Tfap2c transcripts persist in both the inner cell mass and trophectoderm of blastocysts, prompting inquiry into Tfap2c's function in post-lineage establishment. In this study, we delineate the dynamics of TFAP2C during the mouse peri-implantation stage and elucidate its collaboration with the key lineage regulators CDX2 and NANOG. Importantly, we propose that de novo formation of H3K9me3 in the extraembryonic ectoderm during implantation antagonizes TFAP2C binding to crucial developmental genes, thereby maintaining its lineage identity. Together, these results highlight the plasticity of the chromatin environment in designating the genomic binding of highly adaptable lineage-specific TFs and regulating embryonic cell fates.


Subject(s)
CDX2 Transcription Factor , Cell Lineage , Chromatin , Gene Expression Regulation, Developmental , Transcription Factor AP-2 , Animals , Chromatin/metabolism , Mice , Cell Lineage/genetics , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , CDX2 Transcription Factor/metabolism , CDX2 Transcription Factor/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Blastocyst/metabolism , Blastocyst/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Female , Histones/metabolism , Cell Differentiation/genetics , Ectoderm/metabolism , Ectoderm/cytology , Embryonic Development/genetics
6.
Cell Rep Med ; 5(5): 101515, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38631348

ABSTRACT

During pregnancy, germline development is vital for maintaining the continuation of species. Recent studies have shown increased pregnancy risks in COVID-19 patients at the perinatal stage. However, the potential consequence of infection for reproductive quality in developing fetuses remains unclear. Here, we analyze the transcriptome and DNA methylome of the fetal germline following maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We find that infection at early gestational age, a critical period of human primordial germ cell specification and epigenetic reprogramming, trivially affects fetal germ cell (FGC) development. Additionally, FGC-niche communications are not compromised by maternal infection. Strikingly, both general and SARS-CoV-2-specific immune pathways are greatly activated in gonadal niche cells to protect FGCs from maternal infection. Notably, there occurs an "in advance" development tendency in FGCs after maternal infection. Our study provides insights into the impacts of maternal SARS-CoV-2 infection on fetal germline development and serves as potential clinical guidance for future pandemics.


Subject(s)
COVID-19 , Fetus , Germ Cells , SARS-CoV-2 , Humans , Female , COVID-19/virology , COVID-19/immunology , COVID-19/pathology , Pregnancy , Germ Cells/virology , Fetus/virology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/pathology , Gonads/virology , Transcriptome/genetics , Male , DNA Methylation/genetics , Epigenesis, Genetic
7.
Sci China Life Sci ; 67(5): 958-969, 2024 May.
Article in English | MEDLINE | ID: mdl-38305985

ABSTRACT

Vertebrate life begins with fertilization, and then the zygote genome is activated after transient silencing, a process termed zygotic genome activation (ZGA). Despite its fundamental role in totipotency and the initiation of life, the precise mechanism underlying ZGA initiation remains unclear. The existence of minor ZGA implies the possible critical role of noncoding RNAs in the initiation of ZGA. Here, we delineate the expression profile of long noncoding RNAs (lncRNAs) in early mouse embryonic development and elucidate their critical role in minor ZGA. Compared with protein-coding genes (PCGs), lncRNAs exhibit a stronger correlation with minor ZGA. Distinct H3K9me3 profiles can be observed between lncRNA genes and PCGs, and the enrichment of H3K9me3 before ZGA might explain the suspended expression of major ZGA-related PCGs despite possessing PolII pre-configuration. Furthermore, we identified the presence of PolII-enriched MuERV-L around the transcriptional start site of minor ZGA-related lncRNAs, and these repeats are responsible for the activation of minor ZGA-related lncRNAs and subsequent embryo development. Our study suggests that MuERV-L mediates minor ZGA lncRNA activation as a critical driver between epigenetic reprogramming triggered by fertilization and the embryo developmental program, thus providing clues for understanding the regulatory mechanism of totipotency and establishing bona fide totipotent stem cells.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Genome , RNA, Long Noncoding , Zygote , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Zygote/metabolism , Mice , Embryonic Development/genetics , Genome/genetics , Female , Histones/metabolism , Epigenesis, Genetic , Embryo, Mammalian/metabolism
9.
Nat Commun ; 14(1): 4807, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37558707

ABSTRACT

Somatic cell nuclear transfer (SCNT) can be used to reprogram differentiated somatic cells to a totipotent state but has poor efficiency in supporting full-term development. H3K9me3 is considered to be an epigenetic barrier to zygotic genomic activation in 2-cell SCNT embryos. However, the mechanism underlying the failure of H3K9me3 reprogramming during SCNT embryo development remains elusive. Here, we perform genome-wide profiling of H3K9me3 in cumulus cell-derived SCNT embryos. We find redundant H3K9me3 marks are closely related to defective minor zygotic genome activation. Moreover, SCNT blastocysts show severely indistinct lineage-specific H3K9me3 deposition. We identify MAX and MCRS1 as potential H3K9me3-related transcription factors and are essential for early embryogenesis. Overexpression of Max and Mcrs1 significantly benefits SCNT embryo development. Notably, MCRS1 partially rescues lineage-specific H3K9me3 allocation, and further improves the efficiency of full-term development. Importantly, our data confirm the conservation of deficient H3K9me3 differentiation in Sertoli cell-derived SCNT embryos, which may be regulated by alternative mechanisms.


Subject(s)
Histones , Zygote , Histones/genetics , Nuclear Transfer Techniques , Embryonic Development/genetics , Blastocyst , Embryo, Mammalian , Cellular Reprogramming/genetics
10.
Front Cell Infect Microbiol ; 13: 1152198, 2023.
Article in English | MEDLINE | ID: mdl-37216181

ABSTRACT

Prototheca species are the only microalgae known to cause opportunistic infections in vertebrates and humans. Most cases of protothecosis in humans are caused by Prototheca wickerhamii, but knowledge of the pathogenicity and biology of Prototheca is limited. Globally, the diagnostic rate of Prototheca species infection is much lower than the actual rate of P. wickerhamii. The precise mechanisms underlying the pathogenesis of Prototheca infections remain unclear. In this study, we identified a strain of P. wickerhamii with atypical colony morphology. To reveal the morphological differences between P. wickerhamii S1 (mucous) and the molecular basis of its pathogenicity, the transcriptomics, proteomics, and metabolomics of two pathogenic P. wickerhamii strains and one environmental strain were analysed. Interestingly, mannan endo-1,4-ß-mannosidase was significantly downregulated in P. wickerhamii S1, which contributes to a thinner cell wall in S1 compared to strains with typical colony morphology, and the toxicity of macrophages is reduced. Metabolite analysis revealed that the mucoid appearance of P. wickerhamii S1 may have been caused by an increase in linoleic acid, glycerol, and other metabolites. There is still a need to better understand the ecology, aetiology, and pathogenesis of P. wickerhamii, and in particular, its transmission between humans, animals, and the environment, from a One Health perspective.


Subject(s)
One Health , Prototheca , Skin Diseases, Infectious , Animals , Humans , Prototheca/genetics , Proteomics , Transcriptome
11.
Cell Prolif ; 56(9): e13436, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36855927

ABSTRACT

Haploid embryonic stem cells (haESCs) are derived from the inner cell mass of the haploid blastocyst, containing only one set of chromosomes. Extensive and accurate chromatin remodelling occurs during haESC derivation, but the intrinsic transcriptome profiles and chromatin structure of haESCs have not been fully explored. We profiled the transcriptomes, nucleosome positioning, and key histone modifications of four mouse haESC lines, and compared these profiles with those of other closely-related stem cell lines, MII oocytes, round spermatids, sperm, and mouse embryonic fibroblasts. haESCs had transcriptome profiles closer to those of naïve pluripotent stem cells. Consistent with the one X chromosome in haESCs, Xist was repressed, indicating no X chromosome inactivation. haESCs and ESCs shared a similar global chromatin structure. However, a nucleosome depletion region was identified in 2056 promoters in ESCs, which was absent in haESCs. Furthermore, three characteristic spatial relationships were formed between transcription factor motifs and nucleosomes in both haESCs and ESCs, specifically in the linker region, on the nucleosome central surface, and nucleosome borders. Furthermore, the chromatin state of 4259 enhancers was off in haESCs but active in ESCs. Functional annotation of these enhancers revealed enrichment in regulation of the cell cycle, a predominantly reported mechanism of haESC self-diploidization. Notably, the transcriptome profiles and chromatin structure of haESCs were highly preserved during passaging but different from those of differentiated cell types.


Subject(s)
Chromatin , Transcriptome , Animals , Male , Mice , Haploidy , Transcriptome/genetics , Chromatin/genetics , Chromatin/metabolism , Nucleosomes/metabolism , Fibroblasts , Semen , Embryonic Stem Cells
12.
Nat Commun ; 14(1): 1209, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36869051

ABSTRACT

Histone H2A monoubiquitination (H2Aub1) functions as a conserved posttranslational modification in eukaryotes to maintain gene expression and guarantee cellular identity. Arabidopsis H2Aub1 is catalyzed by the core components AtRING1s and AtBMI1s of polycomb repressive complex 1 (PRC1). Because PRC1 components lack known DNA binding domains, it is unclear how H2Aub1 is established at specific genomic locations. Here, we show that the Arabidopsis cohesin subunits AtSYN4 and AtSCC3 interact with each other, and AtSCC3 binds to AtBMI1s. H2Aub1 levels are reduced in atsyn4 mutant or AtSCC3 artificial microRNA knockdown plants. ChIP-seq assays indicate that most binding events of AtSYN4 and AtSCC3 are associated with H2Aub1 along the genome where transcription is activated independently of H3K27me3. Finally, we show that AtSYN4 binds directly to the G-box motif and directs H2Aub1 to these sites. Our study thus reveals a mechanism for cohesin-mediated recruitment of AtBMI1s to specific genomic loci to mediate H2Aub1.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Histones , Polycomb Repressive Complex 1 , Protein Processing, Post-Translational , Ubiquitination , Cohesins
13.
EMBO J ; 42(7): e111112, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36799040

ABSTRACT

Brain metastasis, most commonly originating from lung cancer, increases cancer morbidity and mortality. Although metastatic colonization is the rate-limiting and most complex step of the metastatic cascade, the underlying mechanisms are poorly understood. Here, in vivo genome-wide CRISPR-Cas9 screening revealed that loss of interferon-induced transmembrane protein 1 (IFITM1) promotes brain colonization of human lung cancer cells. Incipient brain metastatic cancer cells with high expression of IFITM1 secrete microglia-activating complement component 3 and enhance the cytolytic activity of CD8+ T cells by increasing the expression and membrane localization of major histocompatibility complex class I. After activation, microglia (of the innate immune system) and cytotoxic CD8+ T lymphocytes (of the adaptive immune system) were found to jointly eliminate cancer cells by releasing interferon-gamma and inducing phagocytosis and T-cell-mediated killing. In human cancer clinical trials, immune checkpoint blockade therapy response was significantly correlated with IFITM1 expression, and IFITM1 enhanced the brain metastasis suppression efficacy of PD-1 blockade in mice. Our results exemplify a novel mechanism through which metastatic cancer cells overcome the innate and adaptive immune responses to colonize the brain, and suggest that a combination therapy increasing IFITM1 expression in metastatic cells with PD-1 blockade may be a promising strategy to reduce metastasis.


Subject(s)
Brain Neoplasms , Lung Neoplasms , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Lung Neoplasms/pathology , Brain/pathology
14.
Adv Sci (Weinh) ; : e2202642, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36382559

ABSTRACT

Lacking a clear understanding of the molecular mechanism determining cancer cell sensitivity to oxidative phosphorylation (OXPHOS) inhibition limits the development of OXPHOS-targeting cancer treatment. Here, cancer cell lines sensitive or resistant to OXPHOS inhibition are identified by screening. OXPHOS inhibition-sensitive cancer cells possess increased OXPHOS activity and silenced nicotinamide N-methyltransferase (NNMT) expression. NNMT expression negatively correlates with OXPHOS inhibition sensitivity and functionally downregulates the intracellular levels of S-adenosyl methionine (SAM). Expression of DNA methyltransferase 1 (DNMT1), a SAM consumer, positively correlates with OXPHOS inhibition sensitivity. NNMT overexpression and DNMT1 inhibition render OXPHOS inhibition-sensitive cancer cells resistant. Importantly, treatments of OXPHOS inhibitors (Gboxin and Berberine) hamper the growth of mouse tumor xenografts by OXPHOS inhibition sensitive but not resistant cancer cells. What's more, the retrospective study of 62 tumor samples from a clinical trial demonstrates that administration of Berberine reduces the tumor recurrence rate of NNMTlow /DNMT1high but not NNMThigh /DNMT1low colorectal adenomas (CRAs). These results thus reveal a critical role of the NNMT-DNMT1 axis in determining cancer cell reliance on mitochondrial OXPHOS and suggest that NNMT and DNMT1 are faithful biomarkers for OXPHOS-targeting cancer therapies.

15.
Cell Rep ; 39(5): 110784, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35508139

ABSTRACT

Assisted reproductive technology has been widely applied in the treatment of human infertility. However, accumulating evidence indicates that in vitro fertilization (IVF) is associated with a low pregnancy rate, placental defects, and metabolic diseases in offspring. Here, we find that IVF manipulation notably disrupts extraembryonic tissue-specific gene expression, and 334 epiblast (Epi)-specific genes and 24 Epi-specific transcription factors are abnormally expressed in extraembryonic ectoderm (ExE) of IVF embryos at embryonic day 7.5. Combined histone modification analysis reveals that aberrant H3K4me3 modification at the Epi active promoters results in increased expression of these genes in ExE. Importantly, we demonstrate that knockdown of the H3K4me3-recruited regulator Kmt2e, which is highly expressed in IVF embryos, greatly improves the development of IVF embryos and reduces abnormal gene expression in ExE. Our study therefore identifies that abnormal H3K4me3 modification in extraembryonic tissue is a major cause of implantation failure and abnormal placental development of IVF embryos.


Subject(s)
Fertilization in Vitro , Placenta , Animals , Female , Germ Layers , Histones , Mice , Placenta/metabolism , Pregnancy , Reproductive Techniques, Assisted
16.
Protein Cell ; 13(8): 580-601, 2022 08.
Article in English | MEDLINE | ID: mdl-35147915

ABSTRACT

Chemically defined medium is widely used for culturing mouse embryonic stem cells (mESCs), in which N2B27 works as a substitution for serum, and GSK3ß and MEK inhibitors (2i) help to promote ground-state pluripotency. However, recent studies suggested that MEKi might cause irreversible defects that compromise the developmental potential of mESCs. Here, we demonstrated the deficient bone morphogenetic protein (BMP) signal in the chemically defined condition is one of the main causes for the impaired pluripotency. Mechanistically, activating the BMP signal pathway by BMP4 could safeguard the chromosomal integrity and proliferation capacity of mESCs through regulating downstream targets Ube2s and Chmp4b. More importantly, BMP4 promotes a distinct in vivo developmental potential and a long-term pluripotency preservation. Besides, the pluripotent improvements driven by BMP4 are superior to those by attenuating MEK suppression. Taken together, our study shows appropriate activation of BMP signal is essential for regulating functional pluripotency and reveals that BMP4 should be applied in the serum-free culture system.


Subject(s)
Bone Morphogenetic Protein 4 , Mouse Embryonic Stem Cells , Pluripotent Stem Cells , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Chromosomal Instability , Endosomal Sorting Complexes Required for Transport , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Signal Transduction , Ubiquitin-Conjugating Enzymes
17.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35210361

ABSTRACT

5-methylcytosine (m5C) is an important epitranscriptomic modification involved in messenger RNA (mRNA) stability and translation efficiency in various biological processes. However, it remains unclear if m5C modification contributes to the dynamic regulation of the transcriptome during the developmental cycles of Plasmodium parasites. Here, we characterize the landscape of m5C mRNA modifications at single nucleotide resolution in the asexual replication stages and gametocyte sexual stages of rodent (Plasmodium yoelii) and human (Plasmodium falciparum) malaria parasites. While different representations of m5C-modified mRNAs are associated with the different stages, the abundance of the m5C marker is strikingly enhanced in the transcriptomes of gametocytes. Our results show that m5C modifications confer stability to the Plasmodium transcripts and that a Plasmodium ortholog of NSUN2 is a major mRNA m5C methyltransferase in malaria parasites. Upon knockout of P. yoelii nsun2 (pynsun2), marked reductions of m5C modification were observed in a panel of gametocytogenesis-associated transcripts. These reductions correlated with impaired gametocyte production in the knockout rodent malaria parasites. Restoration of the nsun2 gene in the knockout parasites rescued the gametocyte production phenotype as well as m5C modification of the gametocytogenesis-associated transcripts. Together with the mRNA m5C profiles for two species of Plasmodium, our findings demonstrate a major role for NSUN2-mediated m5C modifications in mRNA transcript stability and sexual differentiation in malaria parasites.


Subject(s)
5-Methylcytosine/chemistry , Plasmodium falciparum/metabolism , Plasmodium yoelii/growth & development , Plasmodium yoelii/metabolism , Protozoan Proteins/metabolism , RNA, Messenger/metabolism , Germ Cells , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium yoelii/genetics , Transcriptome
18.
Cell Rep ; 37(5): 109912, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731622

ABSTRACT

Fetal growth restriction (FGR) increases the risk for impaired cognitive function later in life. However, the precise mechanisms remain elusive. Using dexamethasone-induced FGR and protein restriction-influenced FGR mouse models, we observe learning and memory deficits in adult FGR offspring. FGR induces decreased hippocampal neurogenesis from the early post-natal period to adulthood by reducing the proliferation of neural stem cells (NSCs). We further find a persistent decrease of Tet1 expression in hippocampal NSCs of FGR mice. Mechanistically, Tet1 downregulation results in hypermethylation of the Dll3 and Notch1 promoters and inhibition of Notch signaling, leading to reduced NSC proliferation. Overexpression of Tet1 activates Notch signaling, offsets the decline in neurogenesis, and enhances learning and memory abilities in FGR offspring. Our data indicate that a long-term decrease in Tet1/Notch signaling in hippocampal NSCs contributes to impaired neurogenesis following FGR and could serve as potential targets for the intervention of FGR-related cognitive disorders.


Subject(s)
Behavior, Animal , Cognition , DNA-Binding Proteins/metabolism , Fetal Growth Retardation/metabolism , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Proto-Oncogene Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/genetics , Disease Models, Animal , Epigenesis, Genetic , Female , Fetal Growth Retardation/physiopathology , Fetal Growth Retardation/psychology , Hippocampus/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory , Mice, Inbred C57BL , Neural Stem Cells/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Proto-Oncogene Proteins/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction
19.
Cell Death Dis ; 12(10): 852, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531373

ABSTRACT

Acute myocardial infarction (AMI), the leading cause of mortality worldwide, is a rapidly developing and irreversible disease. Therefore, proper prompt intervention at the early stage of AMI is crucial for its treatment. However, the molecular features in the early stage have not been clarified. Here, we constructed mouse AMI model and profiled transcriptomes and proteomes at the early stages of AMI progress. Immune system was extensively activated at 6-h AMI. Then, pyroptosis was activated at 24-h AMI. VX-765 treatment, a pyroptosis inhibitor, significantly reduced the infarct size and improved the function of cardiomyocytes. Besides, we identified that WIPI1, specifically expressed in heart, was significantly upregulated at 1 h after AMI. Moreover, WIPI1 expression is significantly higher in the peripheral blood of patients with AMI than healthy control. WIPI1 can serve as a potential early diagnostic biomarker for AMI. It likely decelerates AMI progress by activating autophagy pathways. These findings shed new light on gene expression dynamics in AMI progress, and present a potential early diagnostic marker and a candidate drug for clinical pre-treatment to prolong the optimal cure time.


Subject(s)
Myocardial Infarction/pathology , Pyroptosis , Animals , Autophagy-Related Proteins/metabolism , Biomarkers/metabolism , Dipeptides/pharmacology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Myocardium/immunology , Myocardium/pathology , Myocardium/ultrastructure , Proteome/metabolism , Pyroptosis/drug effects , Pyroptosis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics , para-Aminobenzoates/pharmacology
20.
EMBO J ; 40(21): e107277, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34558085

ABSTRACT

The dorsal and ventral human telencephalons contain different neuronal subtypes, including glutamatergic, GABAergic, and cholinergic neurons, and how these neurons are generated during early development is not well understood. Using scRNA-seq and stringent validations, we reveal here a developmental roadmap for human telencephalic neurons. Both dorsal and ventral telencephalic radial glial cells (RGs) differentiate into neurons via dividing intermediate progenitor cells (IPCs_div) and early postmitotic neuroblasts (eNBs). The transcription factor ASCL1 plays a key role in promoting fate transition from RGs to IPCs_div in both regions. RGs from the regionalized neuroectoderm show heterogeneity, with restricted glutamatergic, GABAergic, and cholinergic differentiation potencies. During neurogenesis, IPCs_div gradually exit the cell cycle and branch into sister eNBs to generate distinct neuronal subtypes. Our findings highlight a general RGs-IPCs_div-eNBs developmental scheme for human telencephalic progenitors and support that the major neuronal fates of human telencephalon are predetermined during dorsoventral regionalization with neuronal diversity being further shaped during neurogenesis and neural circuit integration.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/metabolism , Telencephalon/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Differentiation , Choline/metabolism , Doublecortin Protein/genetics , Doublecortin Protein/metabolism , Fetus , Gene Ontology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Molecular Sequence Annotation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Stathmin/genetics , Stathmin/metabolism , Telencephalon/cytology , Telencephalon/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL