Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 44(1): 48, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822888

ABSTRACT

C3-positive reactive astrocytes play a neurotoxic role in various neurodegenerative diseases. However, the mechanisms controlling C3-positive reactive astrocyte induction are largely unknown. We found that the length of the primary cilium, a cellular organelle that receives extracellular signals was increased in C3-positive reactive astrocytes, and the loss or shortening of primary cilium decreased the count of C3-positive reactive astrocytes. Pharmacological experiments suggested that Ca2+ signalling may synergistically promote C3 expression in reactive astrocytes. Conditional knockout (cKO) mice that specifically inhibit primary cilium formation in astrocytes upon drug stimulation exhibited a reduction in the proportions of C3-positive reactive astrocytes and apoptotic cells in the brain even after the injection of lipopolysaccharide (LPS). Additionally, the novel object recognition (NOR) score observed in the cKO mice was higher than that observed in the neuroinflammation model mice. These results suggest that the primary cilium in astrocytes positively regulates C3 expression. We propose that regulating astrocyte-specific primary cilium signalling may be a novel strategy for the suppression of neuroinflammation.


Subject(s)
Astrocytes , Cilia , Mice, Knockout , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Cilia/metabolism , Cilia/drug effects , Mice , Complement C3/metabolism , Mice, Inbred C57BL , Lipopolysaccharides/pharmacology , Apoptosis/drug effects
2.
Sci Rep ; 14(1): 10636, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724644

ABSTRACT

Gene-knockout animal models with organ-deficient phenotypes used for blastocyst complementation are generally not viable. Animals need to be maintained as heterozygous mutants, and homozygous mutant embryos yield only one-fourth of all embryos. In this study, we generated organ-deficient embryos using the CRISPR-Cas9-sgRNAms system that induces cell death with a single-guide RNA (sgRNAms) targeting multiple sites in the genome. The Cas9-sgRNAms system interrupted cell proliferation and induced cell ablation in vitro. The mouse model had Cas9 driven by the Foxn1 promoter with a ubiquitous expression cassette of sgRNAms at the Rosa26 locus (Foxn1Cas9; Rosa26_ms). It showed an athymic phenotype similar to that of nude mice but was not hairless. Eventually, a rat cell-derived thymus in an interspecies chimera was generated by blastocyst complementation of Foxn1Cas9; Rosa26_ms mouse embryos with rat embryonic stem cells. Theoretically, a half of the total embryos has the Cas9-sgRNAms system because Rosa26_ms could be maintained as homozygous.


Subject(s)
CRISPR-Cas Systems , Forkhead Transcription Factors , RNA, Guide, CRISPR-Cas Systems , Animals , Mice , Rats , RNA, Guide, CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Thymus Gland/metabolism , Models, Animal , Blastocyst/metabolism
3.
Development ; 151(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38179792

ABSTRACT

Regenerative medicine is a tool to compensate for the shortage of lungs for transplantation, but it remains difficult to construct a lung in vitro due to the complex three-dimensional structures and multiple cell types required. A blastocyst complementation method using interspecies chimeric animals has been attracting attention as a way to create complex organs in animals, although successful lung formation using interspecies chimeric animals has not yet been achieved. Here, we applied a reverse-blastocyst complementation method to clarify the conditions required to form lungs in an Fgfr2b-deficient mouse model. We then successfully formed a rat-derived lung in the mouse model by applying a tetraploid-based organ-complementation method. Importantly, rat lung epithelial cells retained their developmental timing even in the mouse body. These findings provide useful insights to overcome the barrier of species-specific developmental timing to generate functional lungs in interspecies chimeras.


Subject(s)
Pluripotent Stem Cells , Rats , Mice , Animals , Receptor, Fibroblast Growth Factor, Type 2/genetics , Blastocyst , Lung , Epithelial Cells , Disease Models, Animal
4.
Sci Rep ; 12(1): 21985, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539541

ABSTRACT

The use of mice as experimental animal models has been a practice since the development of genetically engineered mouse models (GEMMs) in the early 1980s. New technologies, including genome editing, have helped in the time- and cost-efficient generation of GEMMs. However, methods for preparing pseudopregnant females, essential for the generation of GEMMs, remain less advanced. This study proposes a new method to achieve simple production of pseudopregnant female mice using a luteinizing hormone-releasing hormone agonist (LHRHa). A 20 µg LHRHa/mouse was identified as the best dose for inducing estrus synchronization. However, the frequency of copulation was 40% on a single injection. With sequential injections of 20 µg LHRHa/mouse on Days-1 and -2, followed by pairing on Day-5, 74% of LHRHa-treated females copulated with male mice. Moreover, LHRHa treatment did not affect fetal and postnatal development. Eventually, successful generation of offspring via embryo transfer was attained using LHRHa-treated pseudopregnant females. LHRHa administration method is efficient in producing pseudopregnant female mice for the generation of GEMMs, and we expect that it will contribute towards advancing the clinical research.


Subject(s)
Embryo Transfer , Estrus Synchronization , Humans , Pregnancy , Male , Female , Mice , Animals , Prenatal Care , Gonadotropin-Releasing Hormone
5.
Exp Anim ; 71(1): 82-89, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-34544911

ABSTRACT

In mammals, sexual fate is determined by the chromosomes of the male and female gametes during fertilization. Males (XY) or females (XX) are produced when a sperm containing a Y or X-chromosome respectively fertilizes an X-chromosome-containing unfertilized egg. However, sexing of preimplantation stage embryos cannot be conducted visually. To address this, transgenic male mouse models with the ubiquitously expressed green fluorescent protein (GFP) transgene on X- (X-GFP) or Y-chromosomes (Y-GFP) have been established. However, when crossed with wild-type females, sexing of the preimplantation stage embryos by observing the GFP signal is problematic in some cases due to X-inactivation, loss of Y-chromosome (LOY), or loss of transgene fluorescence. In this study, a mouse model with the ubiquitously expressed red fluorescent protein (RFP) transgene on the Y-chromosome was generated since RFP is easily distinguishable from GFP signals. Unfortunately, the ubiquitously expressed tdTomato RFP transgene on the Y-chromosome (Y-RFP) mouse showed the lethal phenotype after birth. No lethal phenotypes were observed when the mitochondrial locating signal N-terminal of tdTomato (mtRFP) was included in the transgene construct. Almost half of the collected fertilized eggs from Y-mtRFP male mice crossed with wild-type females had an RFP signal at the preimplantation stage (E1.5). Therefore, XY eggs were recognized as RFP-positive embryos at the preimplantation stage. Furthermore, 100% sexing was observed at the preimplantation stage using the X-linked GFP/Y-linked RFP male mouse. The established Y-mtRFP mouse models may be used to study sex chromosome related research.


Subject(s)
X Chromosome , Y Chromosome , Animals , Female , Luminescent Proteins , Male , Mice , Mice, Transgenic , X Chromosome/genetics , Y Chromosome/genetics , Red Fluorescent Protein
6.
Sci Rep ; 11(1): 8297, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859300

ABSTRACT

E26 avian leukemia oncogene 2, 3' domain (Ets2) has been implicated in various biological processes. An Ets2 mutant model (Ets2db1/db1), which lacks the DNA-binding domain, was previously reported to exhibit embryonic lethality caused by a trophoblast abnormality. This phenotype could be rescued by tetraploid complementation, resulting in pups with wavy hair and curly whiskers. Here, we generated new Ets2 mutant models with a frame-shift mutation in exon 8 using the CRISPR/Cas9 method. Homozygous mutants could not be obtained by natural mating as embryonic development stopped before E8.5, as previously reported. When we rescued them by tetraploid complementation, these mice did not exhibit wavy hair or curly whisker phenotypes. Our newly generated mice exhibited exon 8 skipping, which led to in-frame mutant mRNA expression in the skin and thymus but not in E7.5 Ets2em1/em1 embryos. This exon 8-skipped Ets2 mRNA was translated into protein, suggesting that this Ets2 mutant protein complemented the Ets2 function in the skin. Our data implies that novel splicing variants incidentally generated after genome editing may complicate the phenotypic analysis but may also give insight into the new mechanisms related to biological gene functions.


Subject(s)
Frameshift Mutation/genetics , Phenotype , Proto-Oncogene Protein c-ets-2/genetics , RNA Splicing/genetics , CRISPR-Cas Systems , DNA-Binding Proteins/genetics , Embryo Loss/genetics , Embryo Loss/pathology , Embryonic Development/genetics , Exons/genetics , Female , Gene Editing/methods , Humans , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/metabolism , Trophoblasts/pathology
7.
Front Cell Dev Biol ; 9: 810118, 2021.
Article in English | MEDLINE | ID: mdl-35096839

ABSTRACT

Fertilization occurs as the culmination of multi-step complex processes. First, mammalian spermatozoa undergo the acrosome reaction to become fusion-competent. Then, the acrosome-reacted spermatozoa penetrate the zona pellucida and adhere to and finally fuse with the egg plasma membrane. IZUMO1 is the first sperm protein proven to be essential for sperm-egg fusion in mammals, as Izumo1 knockout mouse spermatozoa adhere to but fail to fuse with the oolemma. However, the IZUMO1 function in other species remains largely unknown. Here, we generated Izumo1 knockout rats by CRISPR/Cas9 and found the male rats were infertile. Unlike in mice, Izumo1 knockout rat spermatozoa failed to bind to the oolemma. Further investigation revealed that the acrosome-intact sperm binding conceals a decreased number of the acrosome-reacted sperm bound to the oolemma in Izumo1 knockout mice. Of note, we could not see any apparent defects in the binding of the acrosome-reacted sperm to the oolemma in the mice lacking recently found fusion-indispensable genes, Fimp, Sof1, Spaca6, or Tmem95. Collectively, our data suggest that IZUMO1 is required for the sperm-oolemma binding prior to fusion at least in rat.

8.
Biochim Biophys Acta Gene Regul Mech ; 1862(1): 58-70, 2019 01.
Article in English | MEDLINE | ID: mdl-30416088

ABSTRACT

Emerging evidence from recent studies has unraveled the roles of long noncoding RNAs (lncRNAs) in the function of various tissues. However, little is known about the roles of lncRNAs in kidney development. In our present study, we aimed to identify functional lncRNAs in one of the three lineages of kidney progenitor cells, i.e., metanephric mesenchymal (MM) cells. We conducted comprehensive analyses of the chromatin signature and transcriptome by RNA-seq and ChIP-seq. We found seventeen lncRNAs that were expressed specifically in MM cells with an active chromatin signature, while remaining silenced in a bivalent chromatin state in non-MM cells. Out of these MM specific lncRNAs, we identified a lncRNA, Gm29418, in a distal enhancer region of Six2, a key regulatory gene of MM cells. We further identified three transcript variants of Gm29418 by Rapid Amplification of cDNA Ends (RACE), and confirmed that the transcription-start-sites (TSSs) of these variants were consistent with the result of Cap Analysis Gene Expression (CAGE). In support of the enhancer-like function of Gm29418 on Six2 expression, we found that knock-down of Gm29418 by two independent anti-sense locked nucleic acid (LNA) phosphorothioate gapmers suppressed Six2 mRNA expression levels in MM cells. We also found that over-expression of Gm29418 led to an increase in Six2 mRNA expression levels in a mouse MM cell line. In conclusion, we identified a lncRNA, Gm29418, in nephron progenitor cells that has an enhancer-like function on a key regulatory gene, Six2.


Subject(s)
Kidney/growth & development , Nephrons/cytology , RNA, Long Noncoding/physiology , Stem Cells/metabolism , Animals , Chromatin , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mice , Nerve Tissue Proteins/metabolism , Transcriptome
9.
Stem Cell Reports ; 8(2): 401-416, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28089670

ABSTRACT

A method to maintain and rebuild ureteric bud (UB)-like structures from UB cells in vitro could provide a useful tool for kidney regeneration. We aimed in our present study to establish a serum-free culture system that enables the expansion of UB progenitor cells, i.e., UB tip cells, and reconstruction of UB-like structures. We found that fibroblast growth factors or retinoic acid (RA) was sufficient for the survival of UB cells in serum-free condition, while the proliferation and maintenance of UB tip cells required glial cell-derived neurotrophic factor together with signaling from either WNT-ß-catenin pathway or RA. The activation of WNT-ß-catenin signaling in UB cells by endogenous WNT proteins required R-spondins. Together with Rho kinase inhibitor, our culture system facilitated the expansion of UB tip cells to form UB-like structures from dispersed single cells. The UB-like structures thus formed retained the original UB characteristics and integrated into the native embryonic kidneys.


Subject(s)
Kidney/cytology , Kidney/embryology , Morphogenesis , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Cell Survival/drug effects , Fibroblast Growth Factors/metabolism , Mice , Models, Biological , Signal Transduction , Stem Cells/drug effects , Thrombospondins/genetics , Thrombospondins/metabolism , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects
10.
PLoS One ; 10(6): e0129242, 2015.
Article in English | MEDLINE | ID: mdl-26075891

ABSTRACT

Knowledge on how to maintain and expand nephron progenitor cells (NPC) in vitro is important to provide a potentially valuable source for kidney replacement therapies. In our present study, we examined the possibility of optimizing NPC maintenance in the "re-aggregate" system. We found that Six2-expressing (Six2(+))-NPC could be maintained in aggregates reconstituted with dispersed cells from E12.5 mouse embryonic kidneys for at least up to 21 days in culture. The maintenance of Six2(+)-NPC required the presence of ureteric bud cells. The number of Six2(+)-NPC increased by more than 20-fold at day 21, but plateaued after day 14. In an attempt to further sustain NPC proliferation by passage subculture, we found that the new (P1) aggregates reconstituted from the original (P0) aggregates failed to maintain NPC. However, based on the similarity between P1 aggregates and aggregates derived from E15.5 embryonic kidneys, we suspected that the differentiated NPC in P1 aggregates may interfere with NPC maintenance. In support of this notion, we found that preventing NPC differentiation by DAPT, a γ-secretase inhibitor that inhibits Notch signaling pathway, was effective to maintain and expand Six2(+)-NPC in P1 aggregates by up to 65-fold. The Six2(+)-NPC in P1 aggregates retained their potential to epithelialize upon exposure to Wnt signal. In conclusion, we demonstrated in our present study that the "re-aggregation" system can be useful for in vitro maintenance of NPC when combined with γ-secretase inhibitor.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Culture Techniques , Cell Self Renewal/drug effects , Enzyme Inhibitors/pharmacology , Nephrons/cytology , Stem Cells/cytology , Stem Cells/drug effects , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
In Vitro Cell Dev Biol Anim ; 49(7): 479-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23756999

ABSTRACT

Successful derivations of specific neuronal and glial cells from embryonic stem cells have enormous potential for cell therapies and regenerative medicine. However, the low efficiency, the complexity of induction method, and the need for purification represent obstacles that make their application impractical. In this study, we found that PDGFRα(+) cells derived from mouse embryonic stem cells (mESC) can serve as a useful source from which to induce cells that express γ-aminobutyric-acid (GABA)-releasing (GABAergic) neuronal markers. PDGFRα(+) cells were induced from mESC on collagen IV-coated plates in mesenchymal stem cell (MSC) culture medium with limited exposure to retinoic acid, sorted by fluorescence-activated cell sorter and maintained in MSC culture medium containing Y-27632, a Rho-associated kinase inhibitor. We found that supplementation of vascular endothelial growth factor, fibroblast growth factor-basic, and sodium azide (NaN3) to MSC culture medium effectively differentiated PDGFRα(+) cells into cells that express GABAergic neuronal markers, such as Pax2, Dlx2, GAD67 NCAM, and tubulin-ßIII, while markers for oligodendrocyte (Sox2) and astrocyte (Glast) were suppressed. Immunostaining for GABA showed the majority (86 ± 5%) of the induced cells were GABA-positive. We also found that the PDGFRα(+) cells retained such differentiation potential even after more than ten passages and cryopreservation. In summary, this study presents a simple and highly efficient method of inducing cells that express GABAergic neuronal markers from mESC. Together with its ease of maintenance in vitro, PDGFRα(+) cells derived from mESC may serve as a useful source for such purpose.


Subject(s)
Embryonic Stem Cells/cytology , GABAergic Neurons/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers/metabolism , Cell Differentiation , Embryonic Stem Cells/metabolism , GABAergic Neurons/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Oligodendroglia/cytology , Oligodendroglia/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Biochem Biophys Res Commun ; 417(2): 897-902, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22209845

ABSTRACT

The in vitro derivation of renal lineage progenitor cells is essential for renal cell therapy and regeneration. Despite extensive studies in the past, a protocol for renal lineage induction from embryonic stem cells remains unestablished. In this study, we aimed to induce renal lineages from mouse embryonic stem cells (mESC) by following in vivo developmental stages, i.e., the induction of mesoderm (Stage I), intermediate mesoderm (Stage II) and renal lineages (Stage III). For stage I induction, in accordance with known signaling pathways involved in mesoderm development in vivo, i.e., Nodal, bone morphogenic proteins (BMPs) and Wnt, we found that the sequential addition of three factors, i.e., Activin-A (A), a surrogate for Nodal signaling, during days 0-2, A plus BMP-4 (4) during days 2-4, and A4 plus lithium (L), a surrogate for Wnt signaling, during days 4-6, was most effective to induce the mesodermal marker, Brachyury. For stage II induction, the addition of retinoic acid (R) in the continuous presence of A4L during days 6-8 was most effective to induce nephrogenic intermediate mesodermal markers, such as Pax2 and Lim1. Under this condition, more than 30% of cells were stained positive for Pax2, and there was a concomitant decrease in the expression of non-mesodermal markers. For stage III induction, in resemblance to the reciprocal induction between ureteric bud (UB) and metanephric mesenchyme (MM) during kidney development, we found that the exposure to conditioned media derived from UB and MM cells was effective in inducing MM and UB markers, respectively. We also observed the emergence and gradual increase of cell populations expressing progenitor cell marker CD24 from Stage I to Stage III. These CD24(+) cells correlated with higher levels of expression of Brachyury at stage I, Pax2 and Lim1 at stage II and MM markers, such as WT1 and Cadherin 11, after exposure to UB-conditioned media at stage III. In conclusion, our results show that stepwise induction by tracing in vivo developmental stages was effective to generate renal lineage progenitor cells from mESC, and CD24 may serve as a useful surface marker for renal lineage cells at stage II and MM cells at stage III.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Kidney/cytology , Kidney/growth & development , AC133 Antigen , Animals , Antigens, CD/analysis , Antigens, CD/biosynthesis , Bone Morphogenetic Proteins/metabolism , CD24 Antigen/analysis , CD24 Antigen/biosynthesis , Cell Culture Techniques , Cell Line , Cell Tracking , Glycoproteins/analysis , Glycoproteins/biosynthesis , Mesoderm/cytology , Mice , Peptides/analysis , Wnt Proteins/metabolism
13.
Cell Signal ; 23(2): 371-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20940044

ABSTRACT

We found in our present study that lithium (Li(+)) induced the expression of endogenous c-Ret, a tyrosine kinase receptor, in murine inner medullary collecting duct (mIMCD-3) cells. Delineation of the promoter region required for the effect of Li(+) identified a positive regulatory element within 180bp upstream of the transcription initiation site. This region contained three putative GC-rich Sp1 binding sites found to be essential for c-Ret induction by Li(+). The effect of Li(+) was mediated through glycogen synthase kinase 3ß (GSK-3ß) inhibition, although there was no biding site for T cell factor/lymphoid enhancer factor (TCF/LEF) in the 180bp. We found that Li(+) activated the mammalian target of rapamycin (mTOR) pathway via GSK-3ß in these cells, and the effect of Li(+) to induce c-Ret was amenable to the inhibitory effect of the mTOR inhibitor, rapamycin. We also found that alterations in both cellular ß-catenin levels and mTOR activities affected the effect of Li(+) on c-Ret transcription in a cooperative manner. In summary, our results show that Li(+) can induce c-Ret expression in mIMCD-3 cells through both ß-catenin- and mTOR-dependent pathways downstream of GSK-3ß inhibition, which act synergistically on the GC-rich Sp1 binding elements in the promoter region.


Subject(s)
Kidney Tubules, Collecting/drug effects , Lithium Chloride/pharmacology , Proto-Oncogene Proteins c-ret/biosynthesis , Animals , Cell Line , GC Rich Sequence , Gene Expression Regulation , Glycogen Synthase Kinases/antagonists & inhibitors , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mice , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction , TOR Serine-Threonine Kinases/physiology , beta Catenin/physiology
14.
Stem Cells ; 27(4): 796-805, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19350679

ABSTRACT

Sall4 is a mouse homolog of a causative gene of the autosomal dominant disorder Okihiro syndrome. We previously showed that the absence of Sall4 leads to lethality during peri-implantation and that Sall4-null embryonic stem (ES) cells proliferate poorly with intact pluripotency when cultured on feeder cells. Here, we report that, in the absence of feeder cells, Sall4-null ES cells express the trophectoderm marker Cdx2, but are maintained for a long period in an undifferentiated state with minimally affected Oct3/4 expression. Feeder-free Sall4-null ES cells contribute solely to the inner cell mass and epiblast in vivo, indicating that these cells still retain pluripotency and do not fully commit to the trophectoderm. These phenotypes could arise from derepression of the Cdx2 promoter, which is normally suppressed by Sall4 and the Mi2/NuRD HDAC complex. However, proliferation was impaired and G1 phase prolonged in the absence of Sall4, suggesting another role for Sall4 in cell cycle control. Although Sall1, also a Sall family gene, is known to genetically interact with Sall4 in vivo, Sall1-null ES cells have no apparent defects and no exacerbation is observed in ES cells lacking both Sall1 and Sall4, compared with Sall4-null cells. This suggests a unique role for Sall4 in ES cells. Thus, though Sall4 does not contribute to the central machinery of the pluripotency, it stabilizes ES cells by repressing aberrant trophectoderm gene expression.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/physiology , Pluripotent Stem Cells/physiology , Transcription Factors/physiology , Animals , CDX2 Transcription Factor , Cell Cycle/genetics , Embryonic Development/genetics , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...