Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 34: 102060, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38028197

ABSTRACT

Exon-skipping therapy is a promising treatment strategy for Duchenne muscular dystrophy (DMD), which is caused by loss-of-function mutations in the DMD gene encoding dystrophin, leading to progressive cardiomyopathy. In-frame deletion of exons 3-9 (Δ3-9), manifesting a very mild clinical phenotype, is a potential targeted reading frame for exon-skipping by targeting actin-binding domain 1 (ABD1); however, the efficacy of this approach for DMD cardiomyopathy remains uncertain. In this study, we compared three isogenic human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) expressing Δ3-9, frameshifting Δ3-7, or intact DMD. RNA sequencing revealed a resemblance in the expression patterns of mechano-transduction-related genes between Δ3-9 and wild-type samples. Furthermore, we observed similar electrophysiological properties between Δ3-9 and wild-type hiPSC-CMs; Δ3-7 hiPSC-CMs showed electrophysiological alterations with accelerated CaMKII activation. Consistently, Δ3-9 hiPSC-CMs expressed substantial internally truncated dystrophin protein, resulting in maintaining F-actin binding and desmin retention. Antisense oligonucleotides targeting exon 8 efficiently induced skipping exons 8-9 to restore functional dystrophin and electrophysiological parameters in Δ3-7 hiPSC-CMs, bringing the cell characteristics closer to those of Δ3-9 hiPSC-CMs. Collectively, exon-skipping targeting ABD1 to convert the reading frame to Δ3-9 may become a promising therapy for DMD cardiomyopathy.

2.
Stem Cell Res Ther ; 14(1): 240, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679796

ABSTRACT

BACKGROUND: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to treat heart diseases; however, the optimal maturity of hiPSC-CMs for effective regenerative medicine remains unclear. We aimed to investigate the benefits of long-term cultured mature hiPSC-CMs in injured rat hearts. METHODS: Cardiomyocytes were differentiated from hiPSCs via monolayer culturing, and the cells were harvested on day 28 or 56 (D28-CMs or D56-CMs, respectively) after differentiation. We transplanted D28-CMs or D56-CMs into the hearts of rat myocardial infarction models and examined cell retention and engraftment via in vivo bioluminescence imaging and histological analysis. We performed transcriptomic sequencing analysis to elucidate the genetic profiles before and after hiPSC-CM transplantation. RESULTS: Upregulated expression of mature sarcomere genes in vitro was observed in D56-CMs compared with D28-CMs. In vivo bioluminescence imaging studies revealed increased bioluminescence intensity of D56-CMs at 8 and 12 weeks post-transplantation. Histological and immunohistochemical analyses showed that D56-CMs promoted engraftment and maturation in the graft area at 12 weeks post-transplantation. Notably, D56-CMs consistently promoted microvessel formation in the graft area from 1 to 12 weeks post-transplantation. Transcriptomic sequencing analysis revealed that compared with the engrafted D28-CMs, the engrafted D56-CMs enriched genes related to blood vessel regulation at 12 weeks post-transplantation. As shown by transcriptomic and western blot analyses, the expression of a small heat shock protein, alpha-B crystallin (CRYAB), was significantly upregulated in D56-CMs compared with D28-CMs. Endothelial cell migration was inhibited by small interfering RNA-mediated knockdown of CRYAB when co-cultured with D56-CMs in vitro. Furthermore, CRYAB overexpression enhanced angiogenesis in the D28-CM grafts at 4 weeks post-transplantation. CONCLUSIONS: Long-term cultured mature hiPSC-CMs promoted engraftment, maturation and angiogenesis post-transplantation in infarcted rat hearts. CRYAB, which was highly expressed in D56-CMs, was identified as an angiogenic factor from mature hiPSC-CMs. This study revealed the benefits of long-term culture, which may enhance the therapeutic potential of hiPSC-CMs.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Animals , Humans , Rats , Blotting, Western , Cell Differentiation , Cell Movement
3.
Sci Rep ; 11(1): 24199, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921203

ABSTRACT

Germline mutations underlie genetic diversity and species evolution. Previous studies have assessed the theoretical mutation rates and spectra in germ cells mostly by analyzing genetic markers and reporter genes in populations and pedigrees. This study reported the direct measurement of germline mutations by whole-genome sequencing of cultured spermatogonial stem cells in mice, namely germline stem (GS) cells, together with multipotent GS (mGS) cells that spontaneously dedifferentiated from GS cells. GS cells produce functional sperm that can generate offspring by transplantation into seminiferous tubules, whereas mGS cells contribute to germline chimeras by microinjection into blastocysts in a manner similar to embryonic stem cells. The estimated mutation rate of GS and mGS cells was approximately 0.22 × 10-9 and 1.0 × 10-9 per base per cell population doubling, respectively, indicating that GS cells have a lower mutation rate compared to mGS cells. GS and mGS cells also showed distinct mutation patterns, with C-to-T transition as the most frequent in GS cells and C-to-A transversion as the most predominant in mGS cells. By karyotype analysis, GS cells showed recurrent trisomy of chromosomes 15 and 16, whereas mGS cells frequently exhibited chromosomes 1, 6, 8, and 11 amplifications, suggesting that distinct chromosomal abnormalities confer a selective growth advantage for each cell type in vitro. These data provide the basis for studying germline mutations and a foundation for the future utilization of GS cells for reproductive technology and clinical applications.


Subject(s)
Embryonic Stem Cells/metabolism , Genomic Instability/physiology , Animals , Chimera/metabolism , Computational Biology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Male , Mice , Mutation , Reactive Oxygen Species/metabolism , Seminiferous Tubules/metabolism , Spermatogonia/cytology , Spermatozoa
4.
Front Cell Dev Biol ; 9: 615761, 2021.
Article in English | MEDLINE | ID: mdl-33665188

ABSTRACT

Mutations/deficiency of TDRD7, encoding a tudor domain protein involved in post-transcriptional gene expression control, causes early onset cataract in humans. While Tdrd7 is implicated in the control of key lens mRNAs, the impact of Tdrd7 deficiency on microRNAs (miRNAs) and how this contributes to transcriptome misexpression and to cataracts, is undefined. We address this critical knowledge-gap by investigating Tdrd7-targeted knockout (Tdrd7-/-) mice that exhibit fully penetrant juvenile cataracts. We performed Affymetrix miRNA 3.0 microarray analysis on Tdrd7-/- mouse lenses at postnatal day (P) 4, a stage preceding cataract formation. This analysis identifies 22 miRNAs [14 over-expressed (miR-15a, miR-19a, miR-138, miR-328, miR-339, miR-345, miR-378b, miR-384, miR-467a, miR-1224, miR-1935, miR-1946a, miR-3102, miR-3107), 8 reduced (let-7b, miR-34c, miR-298, miR-382, miR-409, miR-1198, miR-1947, miR-3092)] to be significantly misexpressed (fold-change ≥ ± 1.2, p-value < 0.05) in Tdrd7-/- lenses. To understand how these misexpressed miRNAs impact Tdrd7-/- cataract, we predicted their mRNA targets and examined their misexpression upon Tdrd7-deficiency by performing comparative transcriptomics analysis on P4 and P30 Tdrd7-/- lens. To prioritize these target mRNAs, we used various stringency filters (e.g., fold-change in Tdrd7-/- lens, iSyTE-based lens-enriched expression) and identified 98 reduced and 89 elevated mRNA targets for overexpressed and reduced miRNAs, respectively, which were classified as "top-priority" "high-priority," and "promising" candidates. For Tdrd7-/- lens overexpressed miRNAs, this approach identified 18 top-priority reduced target mRNAs: Alad, Ankrd46, Ceacam10, Dgat2, Ednrb, H2-Eb1, Klhl22, Lin7a, Loxl1, Lpin1, Npc1, Olfm1, Ppm1e, Ppp1r1a, Rgs8, Shisa4, Snx22 and Wnk2. Majority of these targets were also altered in other gene-specific perturbation mouse models (e.g., Brg1, E2f1/E2f2/E2f3, Foxe3, Hsf4, Klf4, Mafg/Mafk, Notch) of lens defects/cataract, suggesting their importance to lens biology. Gene ontology (GO) provided further insight into their relevance to lens pathology. For example, the Tdrd7-deficient lens capsule defect may be explained by reduced mRNA targets (e.g., Col4a3, Loxl1, Timp2, Timp3) associated with "basement membrane". GO analysis also identified new genes (e.g., Casz1, Rasgrp1) recently linked to lens biology/pathology. Together, these analyses define a new Tdrd7-downstream miRNA-mRNA network, in turn, uncovering several new mRNA targets and their associated pathways relevant to lens biology and offering molecular insights into the pathology of congenital cataract.

5.
Sci Rep ; 10(1): 11883, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681032

ABSTRACT

Accumulating evidence suggests that human pluripotent stem cell-derived cardiomyocytes can affect "heart regeneration", replacing injured cardiac scar tissue with concomitant electrical integration. However, electrically coupled graft cardiomyocytes were found to innately induce transient post-transplant ventricular tachycardia in recent large animal model transplantation studies. We hypothesised that these phenomena were derived from alterations in the grafted cardiomyocyte characteristics. In vitro experiments showed that human embryonic stem cell-derived cardiomyocytes (hESC-CMs) contain nodal-like cardiomyocytes that spontaneously contract faster than working-type cardiomyocytes. When transplanted into athymic rat hearts, proliferative capacity was lower for nodal-like than working-type cardiomyocytes with grafted cardiomyocytes eventually comprising only relatively matured ventricular cardiomyocytes. RNA-sequencing of engrafted hESC-CMs confirmed the increased expression of matured ventricular cardiomyocyte-related genes, and simultaneous decreased expression of nodal cardiomyocyte-related genes. Temporal engraftment of electrical excitable nodal-like cardiomyocytes may thus explain the transient incidence of post-transplant ventricular tachycardia, although further large animal model studies will be required to control post-transplant arrhythmia.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Regeneration , Action Potentials , Biomarkers , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Phylogeny
6.
Hum Mol Genet ; 29(12): 2076-2097, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32420594

ABSTRACT

Mutations of the RNA granule component TDRD7 (OMIM: 611258) cause pediatric cataract. We applied an integrated approach to uncover the molecular pathology of cataract in Tdrd7-/- mice. Early postnatal Tdrd7-/- animals precipitously develop cataract suggesting a global-level breakdown/misregulation of key cellular processes. High-throughput RNA sequencing integrated with iSyTE-bioinformatics analysis identified the molecular chaperone and cytoskeletal modulator, HSPB1, among high-priority downregulated candidates in Tdrd7-/- lens. A protein fluorescence two-dimensional difference in-gel electrophoresis (2D-DIGE)-coupled mass spectrometry screen also identified HSPB1 downregulation, offering independent support for its importance to Tdrd7-/- cataractogenesis. Lens fiber cells normally undergo nuclear degradation for transparency, posing a challenge: how is their cell morphology, also critical for transparency, controlled post-nuclear degradation? HSPB1 functions in cytoskeletal maintenance, and its reduction in Tdrd7-/- lens precedes cataract, suggesting cytoskeletal defects may contribute to Tdrd7-/- cataract. In agreement, scanning electron microscopy (SEM) revealed abnormal fiber cell morphology in Tdrd7-/- lenses. Further, abnormal phalloidin and wheat germ agglutinin (WGA) staining of Tdrd7-/- fiber cells, particularly those exhibiting nuclear degradation, reveals distinct regulatory mechanisms control F-actin cytoskeletal and/or membrane maintenance in post-organelle degradation maturation stage fiber cells. Indeed, RNA immunoprecipitation identified Hspb1 mRNA in wild-type lens lysate TDRD7-pulldowns, and single-molecule RNA imaging showed co-localization of TDRD7 protein with cytoplasmic Hspb1 mRNA in differentiating fiber cells, suggesting that TDRD7-ribonucleoprotein complexes may be involved in optimal buildup of key factors. Finally, Hspb1 knockdown in Xenopus causes eye/lens defects. Together, these data uncover TDRD7's novel upstream role in elevation of stress-responsive chaperones for cytoskeletal maintenance in post-nuclear degradation lens fiber cells, perturbation of which causes early-onset cataracts.


Subject(s)
Cataract/genetics , Eye Proteins/genetics , Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Ribonucleoproteins/genetics , Animals , Cataract/pathology , Cell Nucleus/genetics , Cytoskeleton/genetics , Disease Models, Animal , Eye Diseases , Humans , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Mice , Microscopy, Electron, Scanning , Mutation/genetics , RNA, Messenger/genetics , Xenopus laevis/genetics
7.
Dev Cell ; 52(4): 429-445.e10, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32032549

ABSTRACT

The mechanisms regulating meiotic initiation in mammals are enigmatic. It is known that retinoic acid (RA) signaling plays a pivotal role during meiotic initiation. STRA8, which is expressed in response to RA, is thought to be a key factor promoting meiotic initiation. However, the specific role of STRA8 in meiotic initiation has remained elusive. Here, we identified MEIOSIN as a germ-cell-specific factor that associates with STRA8. MEIOSIN, like STRA8, is expressed in response to RA and plays an essential role in meiotic initiation in both males and females. Functional analyses revealed that MEIOSIN acts as a transcription factor together with STRA8, and that both factors are critical for driving meiotic gene activation. Furthermore, temporally restricted expression of MEIOSIN leads to meiotic entry decision during spermatogenesis. The present study demonstrates that MEIOSIN, in collaboration with STRA8, plays a central role in regulating the mitosis to meiosis germ cell fate decision in mammals.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Cycle , Gene Expression Regulation , Germ Cells/physiology , Meiosis , Mitosis , Transcription Factors/physiology , Animals , Cell Differentiation , Female , Germ Cells/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Spermatogenesis
8.
Stem Cell Reports ; 14(3): 506-519, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32084386

ABSTRACT

Naive and primed human pluripotent stem cells (hPSCs) have provided useful insights into the regulation of pluripotency. However, the molecular mechanisms regulating naive conversion remain elusive. Here, we report intermediate naive conversion induced by overexpressing nuclear receptor 5A1 (NR5A1) in hPSCs. The cells displayed some naive features, such as clonogenicity, glycogen synthase kinase 3ß, and mitogen-activated protein kinase (MAPK) independence, expression of naive-associated genes, and two activated X chromosomes, but lacked others, such as KLF17 expression, transforming growth factor ß independence, and imprinted gene demethylation. Notably, NR5A1 negated MAPK activation by fibroblast growth factor 2, leading to cell-autonomous self-renewal independent of MAPK inhibition. These phenotypes may be associated with naive conversion, and were regulated by a DPPA2/4-dependent pathway that activates the selective expression of naive-associated genes. This study increases our understanding of the mechanisms regulating the conversion from primed to naive pluripotency.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Steroidogenic Factor 1/metabolism , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Histones/metabolism , Humans , Principal Component Analysis , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Steroidogenic Factor 1/genetics , Transcription, Genetic/drug effects
9.
Biol Reprod ; 101(1): 248-256, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30951587

ABSTRACT

PIWI-interacting RNAs (piRNAs), a subclass of germ cell-specific noncoding small RNAs, are essential for de novo DNA methylation of retrotransposon genes in embryonic testes. PIWIL2/MILI, one of three mouse PIWI family members, is indispensable for piRNA production, DNA methylation of retrotransposons presumably via piRNA, and normal spermatogenesis. In vitro analysis using germline stem cells (GS cells) revealed that glycerol-3-phosphate acyltransferase 2 (GPAT2), which is a mitochondrial outer membrane protein involved in generation of lysophosphatidic acid (LPA) and highly expressed in testes, plays important roles in spermatogenesis. Namely, GPAT2 binds to PIWIL2 and is closely involved in the biogenesis of piRNAs; this process is independent of its enzymatic activity on LPA. However, GS cells recapitulate only a limited phase of spermatogenesis and the biological functions of GPAT2 remain largely unknown. In this study, we generated GPAT2-deficient mice and conducted comprehensive analyses. The deficient mice showed defective piRNA production and subsequent de-silencing of IAP and Line-1 retrotransposons in fetal testes. In addition, apoptosis of pachytene spermatocytes was observed. These abnormalities were all common to the phenotype of PIWIL2-deficient mice, in which piRNA production was impaired. GPAT2-deficient mice exhibited apoptosis in spermatogonia at the neonatal stage, which was not observed in PIWIL2-deficient mice. These data show that GPAT2 plays a critical role in preventing apoptosis in spermatogonia.


Subject(s)
Gene Silencing/physiology , Glycerol-3-Phosphate O-Acyltransferase/physiology , RNA, Small Interfering/biosynthesis , Retroelements/genetics , Spermatogonia/physiology , Animals , Cell Proliferation/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Glycerol-3-Phosphate O-Acyltransferase/genetics , Male , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Spermatogenesis/genetics , Spermatogonia/cytology , Testis/cytology , Testis/metabolism
10.
EMBO Rep ; 19(4)2018 04.
Article in English | MEDLINE | ID: mdl-29437694

ABSTRACT

The piRNA pathway is a piRNA-guided retrotransposon silencing system which includes processing of retrotransposon transcripts by PIWI-piRNAs in secondary piRNA biogenesis. Although several proteins participate in the piRNA pathway, the ones crucial for the cleavage of target RNAs by PIWI-piRNAs have not been identified. Here, we show that GTSF1, an essential factor for retrotransposon silencing in male germ cells in mice, associates with both MILI and MIWI2, mouse PIWI proteins that function in prospermatogonia. GTSF1 deficiency leads to a severe defect in the production of secondary piRNAs, which are generated from target RNAs of PIWI-piRNAs. Furthermore, in Gtsf1 mutants, a known target RNA of PIWI-piRNAs is left unsliced at the cleavage site, and the generation of secondary piRNAs from this transcript is defective. Our findings indicate that GTSF1 is a crucial factor for the slicing of target RNAs by PIWI-piRNAs and thus affects secondary piRNA biogenesis in prospermatogonia.


Subject(s)
Gene Expression Regulation , Proteins/metabolism , RNA, Small Interfering/genetics , Transcription, Genetic , Adult Germline Stem Cells/metabolism , Animals , Cell Nucleus/metabolism , Gene Amplification , Gene Silencing , Genes, Intracisternal A-Particle , Intracellular Signaling Peptides and Proteins , Long Interspersed Nucleotide Elements , Male , Mice , Mice, Knockout , Models, Biological , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , Proteins/genetics , RNA Interference , Recombinant Fusion Proteins , Retroelements , Testis/metabolism
11.
Mol Cell Biol ; 37(23)2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28894029

ABSTRACT

DNA replication is frequently perturbed by intrinsic, as well as extrinsic, genotoxic stress. At damaged forks, DNA replication and repair activities require proper coordination to maintain genome integrity. We show here that PARI antirecombinase plays an essential role in modulating the initial response to replication stress in mice. PARI is functionally dormant at replisomes during normal replication, but upon replication stress, it enhances nascent-strand shortening that is regulated by RAD51 and MRE11. PARI then promotes double-strand break induction, followed by new origin firing instead of replication restart. Such PARI function is apparently obstructive to replication but is nonetheless physiologically required for chromosome stability in vivo and ex vivo Of note, Pari-deficient embryonic stem cells exhibit spontaneous chromosome instability, which is attenuated by differentiation induction, suggesting that pluripotent stem cells have a preferential requirement for PARI that acts against endogenous replication stress. PARI is a latent modulator of stalled fork processing, which is required for stable genome inheritance under both endogenous and exogenous replication stress in mice.


Subject(s)
Chromosomal Instability/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Genomic Instability/genetics , Animals , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Humans , Mice
12.
Nucleic Acids Res ; 45(9): 5387-5398, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28115634

ABSTRACT

The mouse PIWI-interacting RNA (piRNA) pathway produces a class of 26-30-nucleotide (nt) small RNAs and is essential for spermatogenesis and retrotransposon repression. In oocytes, however, its regulation and function are poorly understood. In the present study, we investigated the consequences of loss of piRNA-pathway components in growing oocytes. When MILI (or PIWIL2), a PIWI family member, was depleted by gene knockout, almost all piRNAs disappeared. This severe loss of piRNA was accompanied by an increase in transcripts derived from specific retrotransposons, especially IAPs. MIWI (or PIWIL1) depletion had a smaller effect. In oocytes lacking PLD6 (or ZUCCHINI or MITOPLD), a mitochondrial nuclease/phospholipase involved in piRNA biogenesis in male germ cells, the piRNA level was decreased to 50% compared to wild-type, a phenotype much milder than that in males. Since PLD6 is essential for the creation of the 5΄ ends of primary piRNAs in males, the presence of mature piRNA in PLD6-depleted oocytes suggests the presence of compensating enzymes. Furthermore, we identified novel 21-23-nt small RNAs, termed spiRNAs, possessing a 10-nt complementarity with piRNAs, which were produced dependent on MILI and independent of DICER. Our study revealed the differences in the biogenesis and function of the piRNA pathway between sexes.


Subject(s)
Argonaute Proteins/metabolism , Mitochondrial Proteins/metabolism , Oocytes/cytology , Oocytes/metabolism , Phospholipase D/metabolism , Animals , Cell Proliferation , Female , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Mice, Inbred C57BL , Oocytes/ultrastructure , Ovary/metabolism , RNA, Small Interfering/metabolism , Retroelements/genetics
13.
Biochem Biophys Res Commun ; 478(2): 592-8, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27473657

ABSTRACT

D1Pas1 is a mouse autosomal DEAD-box RNA helicase expressed predominantly in the testis. To assess its possible function, we generated D1Pas1-deficient mice using embryonic stem cells with a targeted D1Pas1 allele. Deletion of D1Pas1 did not cause noticeable embryonic defects or death, indicating that D1Pas1 is not essential for embryogenesis. Whereas homozygous knockout female mice showed normal reproductive performance, homozygous knockout male mice were completely sterile. The seminiferous epithelium of D1Pas1-deficient males contained no spermatids or spermatozoa because of spermatogenic arrest at the late pachytene stage. Upregulation of retrotransposons such as LINE-1 was not found in D1Pas1-deficient males, unlike males lacking Mvh, another testicular DEAD-box RNA helicase. Meiotic chromosome behavior in developing spermatocytes of D1Pas1-deficient males was indistinguishable from that in wild-type males, at least until synaptonemal complex formation. Thus, mouse D1Pas1 is the first-identified DEAD-box RNA helicase that plays critical roles in the final step of the first meiotic prophase in male germ cells.


Subject(s)
DEAD-box RNA Helicases/genetics , Meiosis , Spermatogenesis , Animals , DEAD-box RNA Helicases/metabolism , Female , Gene Knockout Techniques , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retroelements , Spermatocytes/cytology , Spermatocytes/metabolism
14.
Nucleic Acids Res ; 42(19): 11903-11, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25262350

ABSTRACT

HSP90, found in all kingdoms of life, is a major chaperone protein regulating many client proteins. We demonstrated that HSP90α, one of two paralogs duplicated in vertebrates, plays an important role in the biogenesis of fetal PIWI-interacting RNAs (piRNA), which act against the transposon activities, in mouse male germ cells. The knockout mutation of Hsp90α resulted in a large reduction in the expression of primary and secondary piRNAs and mislocalization of MIWI2, a PIWI homolog. Whereas the mutation in Fkbp6 encoding a co-chaperone reduced piRNAs of 28-32 nucleotides in length, the Hsp90α mutation reduced piRNAs of 24-32 nucleotides, suggesting the presence of both FKBP6-dependent and -independent actions of HSP90α. Although DNA methylation and mRNA levels of L1 retrotransposon were largely unchanged in the Hsp90α mutant testes, the L1-encoded protein was increased, suggesting the presence of post-transcriptional regulation. This study revealed the specialized function of the HSP90α isofom in the piRNA biogenesis and repression of retrotransposons during the development of male germ cells in mammals.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , RNA, Small Interfering/metabolism , Retroelements , Animals , Arginine/metabolism , Argonaute Proteins/analysis , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Fetus/metabolism , HSP90 Heat-Shock Proteins/genetics , Male , Methylation , Mice , Mice, Knockout , Mutation , Testis/embryology , Testis/metabolism
15.
Dev Cell ; 29(5): 501-502, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24914555

ABSTRACT

Fetal oocytes in mammals undergo extensive apoptosis during development. In this issue of Developmental Cell, Malki et al. (2014) provide insight into how and why such massive oocyte loss occurs through the demonstration that the expression level of LINE-1 retrotransposon defines the survival threshold and thus viability of fetal oocytes.


Subject(s)
DNA-Binding Proteins/physiology , Endodeoxyribonucleases/physiology , Fetus/cytology , Long Interspersed Nucleotide Elements/genetics , Meiosis/physiology , Oocytes/cytology , Ovary/cytology , Transcription Factors/physiology , Animals , Female , Male
16.
Proc Natl Acad Sci U S A ; 110(41): 16492-7, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24067652

ABSTRACT

Piwi-interacting RNAs (piRNAs) are gonad-specific small RNAs that provide defense against transposable genetic elements called transposons. Our knowledge of piRNA biogenesis is sketchy, partly due to an incomplete inventory of the factors involved. Here, we identify Tudor domain-containing 12 (TDRD12; also known as ECAT8) as a unique piRNA biogenesis factor in mice. TDRD12 is detected in complexes containing Piwi protein MILI (PIWIL2), its associated primary piRNAs, and TDRD1, all of which are already implicated in secondary piRNA biogenesis. Male mice carrying either a nonsense point mutation (reproductive mutant 23 or repro23 mice) or a targeted deletion in the Tdrd12 locus are infertile and derepress retrotransposons. We find that TDRD12 is dispensable for primary piRNA biogenesis but essential for production of secondary piRNAs that enter Piwi protein MIWI2 (PIWIL4). Cell-culture studies with the insect ortholog of TDRD12 suggest a role for the multidomain protein in mediating complex formation with other participants during secondary piRNA biogenesis.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Methylation/physiology , DNA Transposable Elements/physiology , Germ Cells/physiology , RNA, Small Interfering/biosynthesis , RNA-Induced Silencing Complex/physiology , Amino Acid Sequence , Animals , Blotting, Northern , Bombyx , Cloning, Molecular , Computational Biology , DNA Primers/genetics , DNA Transposable Elements/genetics , DNA, Complementary/genetics , Fluorescent Antibody Technique , Genetic Vectors/genetics , Immunoprecipitation , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA-Induced Silencing Complex/genetics
17.
Development ; 140(18): 3819-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23924633

ABSTRACT

Mobilization of endogenous retrotransposons can destabilize the genome, an imminent danger during epigenetic reprogramming of cells in the germline. The P-element-induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway is known to silence retrotransposons in the mouse testes. Several piRNA pathway components localize to the unique, germline structure known as the nuage. In this study, we surveyed mouse ovaries and found, for the first time, transient appearance of nuage-like structures in oocytes of primordial follicles. Mouse vasa homolog (MVH), Piwi-like 2 (PIWIL2/MILI) and tudor domain-containing 9 (TDRD9) are present in these structures, whereas aggregates of germ cell protein with ankyrin repeats, sterile alpha motif and leucine zipper (GASZ) localize separately in the cytoplasm. Retrotransposons are silenced in primordial ovarian follicles, and de-repressed upon reduction of piRNA expression in Mvh, Mili or Gasz mutants. However, these null-mutant females, unlike their male counterparts, are fertile, uncoupling retrotransposon activation from sterility.


Subject(s)
Cellular Structures/metabolism , Gene Silencing , Ovarian Follicle/metabolism , Retroelements/genetics , Animals , Cellular Structures/ultrastructure , Female , Gene Expression Regulation , Germ Cells/metabolism , Infertility, Female/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Oogenesis , Ovarian Follicle/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
18.
RNA ; 19(6): 803-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23611983

ABSTRACT

piRNA (PIWI-interacting RNA) is a germ cell-specific small RNA in which biogenesis PIWI (P-element wimpy testis) family proteins play crucial roles. MILI (mouse Piwi-like), one of the three mouse PIWI family members, is indispensable for piRNA production, DNA methylation of retrotransposons presumably through the piRNA, and spermatogenesis. The biogenesis of piRNA has been divided into primary and secondary processing pathways; in both of these MILI is involved in mice. To analyze the molecular function of MILI in piRNA biogenesis, we utilized germline stem (GS) cells, which are derived from testicular stem cells and possess a spermatogonial phenotype. We established MILI-null GS cell lines and their revertant, MILI-rescued GS cells, by introducing the Mili gene with Sendai virus vector. Comparison of wild-type, MILI-null, and MILI-rescued GS cells revealed that GS cells were quite useful for analyzing the molecular mechanisms of piRNA production, especially the primary processing pathway. We found that glycerol-3-phosphate acyltransferase 2 (GPAT2), a mitochondrial outer membrane protein for lysophosphatidic acid, bound to MILI using the cells and that gene knockdown of GPAT2 brought about impaired piRNA production in GS cells. GPAT2 is not only one of the MILI bound proteins but also a protein essential for primary piRNA biogenesis.


Subject(s)
Glycerol-3-Phosphate O-Acyltransferase/metabolism , RNA, Small Interfering/metabolism , Stem Cells/metabolism , Testis/metabolism , Animals , Animals, Newborn , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Blotting, Western , Cell Cycle Proteins , Cells, Cultured , Gene Knockdown Techniques , Genetic Vectors/metabolism , Glycerol-3-Phosphate O-Acyltransferase/genetics , Immunoprecipitation , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred DBA , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Protein Binding , RNA, Small Interfering/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Sendai virus/genetics , Sendai virus/metabolism , Stem Cells/cytology , Testis/cytology
19.
Philos Trans R Soc Lond B Biol Sci ; 368(1609): 20110338, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23166399

ABSTRACT

Transposable elements and their fossil sequences occupy about half of the genome in mammals. While most of these selfish mobile elements have been inactivated by truncations and mutations during evolution, some copies remain competent to transpose and/or amplify, posing an ongoing genetic threat. To control such mutagenic sequences, host genomes have developed multiple layers of defence mechanisms, including epigenetic regulation and RNA silencing. Germ cells, in particular, employ the piwi-small RNA pathway, which plays a central and adaptive role in safeguarding the germline genome from retrotransposons. Recent studies have revealed that a class of developmentally regulated genes, which have long been implicated in germ cell specification and differentiation, such as vasa and tudor family genes, play key roles in the piwi pathway to suppress retrotransposons, indicating that the piwi-mediated genome protection is at the core of germline development. Furthermore, while the piwi system primarily operates post-transcriptionally at the RNA level, it also affects the epigenetics of cognate genome loci, offering an intriguing link between small RNAs and transcriptional control in mammals. In this review, we summarize our current understanding of the piwi pathway in mice, which is emerging as a fundamental component of spermatogenesis that ensures male fertility and genome integrity.


Subject(s)
Fertility/genetics , RNA, Small Interfering/metabolism , Spermatogenesis , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Cycle Proteins , DNA Methylation , Epigenesis, Genetic , Male , Meiosis , Mice , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Retroelements , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism
20.
Mol Cell ; 47(6): 970-9, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22902560

ABSTRACT

Epigenetic silencing of transposons by Piwi-interacting RNAs (piRNAs) constitutes an RNA-based genome defense mechanism. Piwi endonuclease action amplifies the piRNA pool by generating new piRNAs from target transcripts by a poorly understood mechanism. Here, we identified mouse Fkbp6 as a factor in this biogenesis pathway delivering piRNAs to the Piwi protein Miwi2. Mice lacking Fkbp6 derepress LINE1 (L1) retrotransposon and display reduced DNA methylation due to deficient nuclear accumulation of Miwi2. Like other cochaperones, Fkbp6 associates with the molecular chaperone Hsp90 via its tetratricopeptide repeat (TPR) domain. Inhibition of the ATP-dependent Hsp90 activity in an insect cell culture model results in the accumulation of short antisense RNAs in Piwi complexes. We identify these to be byproducts of piRNA amplification that accumulate only in nuage-localized Piwi proteins. We propose that the chaperone machinery normally ejects these inhibitory RNAs, allowing turnover of Piwi complexes for their continued participation in piRNA amplification.


Subject(s)
Long Interspersed Nucleotide Elements , RNA Interference , RNA, Small Interfering/genetics , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Animals , Argonaute Proteins/biosynthesis , Argonaute Proteins/metabolism , Cell Line , DNA Methylation , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Knockout , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Tacrolimus Binding Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...