Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Exp Anim ; 72(4): 460-467, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37183025

ABSTRACT

Adeno-associated virus serotype 9 (AAV9) has become a popular tool for gene transfer because of its ability to cross the blood-brain barrier and efficiently transduce genetic material into a variety of cell types. The study utilized GRR (Green-to-Red Reporter) mouse embryos, in which the expression of iCre results in the disappearance of Green Fluorescent Protein (GFP) expression and the detection of Discosoma sp. Red Fluorescent Protein (DsRed) expression by intraplacental injection. Our results demonstrate that AAV9-CMV-iCre can transduce multiple organs in embryos at developmental stages E9.5-E11.5, including the liver, heart, brain, thymus, and intestine. These findings suggest that intraplacental injection of AAV9-CMV-iCre is a viable method for the widespread transduction of GRR mouse embryos.


Subject(s)
Cytomegalovirus Infections , Dependovirus , Mice , Animals , Dependovirus/genetics , Serogroup , Brain/metabolism , Blood-Brain Barrier , Cytomegalovirus Infections/metabolism , Genetic Vectors , Transduction, Genetic
2.
J Reprod Dev ; 69(1): 48-52, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36529517

ABSTRACT

We examined various methods to enhance the accessibility of intracytoplasmic sperm injection (ICSI) technology to more users by making the technique easier, more efficient, and practical. First, the methods for artificially removing the mouse sperm tail were evaluated. Trypsin treatment was found to efficiently remove the sperm tails. The resultant sperm cells had a lower oocyte activation capacity; however, the use of activated oocytes resulted in the same fecundity as that of fresh, untreated sperm. Pre-activated oocytes were more resistant to physical damage, showed higher survival rates, and required less time per injection. Testing this method in rats yielded similar results, although the oocyte activation method was different. Remarkably, this method resulted in higher birth rates of rat progeny than with conventional methods of rat ICSI. Our method thereby streamlines mouse and rat ICSI, making it more accessible to laboratories across many disciplines.


Subject(s)
Sperm Injections, Intracytoplasmic , Sperm Tail , Mice , Male , Rats , Animals , Sperm Injections, Intracytoplasmic/methods , Trypsin , Semen , Spermatozoa/physiology , Oocytes
3.
Sci Rep ; 11(1): 9168, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33911096

ABSTRACT

Spaceflight causes a decrease in skeletal muscle mass and strength. We set two murine experimental groups in orbit for 35 days aboard the International Space Station, under artificial earth-gravity (artificial 1 g; AG) and microgravity (µg; MG), to investigate whether artificial 1 g exposure prevents muscle atrophy at the molecular level. Our main findings indicated that AG onboard environment prevented changes under microgravity in soleus muscle not only in muscle mass and fiber type composition but also in the alteration of gene expression profiles. In particular, transcriptome analysis suggested that AG condition could prevent the alterations of some atrophy-related genes. We further screened novel candidate genes to reveal the muscle atrophy mechanism from these gene expression profiles. We suggest the potential role of Cacng1 in the atrophy of myotubes using in vitro and in vivo gene transductions. This critical project may accelerate the elucidation of muscle atrophy mechanisms.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/physiology , Muscular Atrophy/genetics , Weightlessness , Adaptation, Biological/genetics , Animals , Calcium Channels/genetics , Cell Line , Gene Expression Profiling , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiopathology , Space Flight
4.
Sci Rep ; 11(1): 4374, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33623082

ABSTRACT

In order to increase the contribution of donor HSC cells, irradiation and DNA alkylating agents have been commonly used as experimental methods to eliminate HSCs for adult mice. But a technique of HSC deletion for mouse embryo for increase contribution of donor cells has not been published. Here, we established for the first time a procedure for placental HSC transplantation into E11.5 Runx1-deficient mice mated with G1-HRD-Runx1 transgenic mice (Runx1-/-::Tg mice) that have no HSCs in the fetal liver. Following the transplantation of fetal liver cells from mice (allogeneic) or rats (xenogeneic), high donor cell chimerism was observed in Runx1-/-::Tg embryos. Furthermore, chimerism analysis and colony assay data showed that donor fetal liver hematopoietic cells contributed to both white blood cells and red blood cells. Moreover, secondary transplantation into adult recipient mice indicated that the HSCs in rescued Runx1-/-::Tg embryos had normal abilities. These results suggest that mice lacking fetal liver HSCs are a powerful tool for hematopoiesis reconstruction during the embryonic stage and can potentially be used in basic research on HSCs or xenograft models.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cell Transplantation/methods , Placenta/cytology , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Rats , Transplantation, Heterologous/methods , Transplantation, Homologous/methods
5.
Kidney Int ; 98(2): 391-403, 2020 08.
Article in English | MEDLINE | ID: mdl-32622525

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a common cause of steroid-resistant nephrotic syndrome. Spontaneous remission of FSGS is rare and steroid-resistant FSGS frequently progresses to renal failure. Many inheritable forms of FSGS have been described, caused by mutations in proteins that are important for podocyte function. Here, we show that a basic leucine zipper transcription factor, MafB, protects against FSGS. MAFB expression was found to be decreased in the podocytes of patients with FSGS. Moreover, conditional podocyte-specific MafB-knockout mice developed FSGS with massive proteinuria accompanied by depletion of the slit diaphragm-related proteins (Nphs1 and Magi2), and the podocyte-specific transcription factor Tcf21. These findings indicate that MafB plays a crucial role in the pathogenesis of FSGS. Consistent with this, adriamycin-induced FSGS and attendant proteinuria were ameliorated by MafB overexpression in the podocytes of MafB podocyte-specific transgenic mice. Thus, MafB could be a new therapeutic target for FSGS.


Subject(s)
Glomerulosclerosis, Focal Segmental , Nephrotic Syndrome , Podocytes , Animals , Basic Helix-Loop-Helix Transcription Factors , Glomerulosclerosis, Focal Segmental/genetics , Humans , MafB Transcription Factor/genetics , Mice , Mice, Transgenic , Nephrotic Syndrome/genetics , Proteinuria/genetics , Proteinuria/prevention & control
6.
Biochem Biophys Res Commun ; 523(2): 452-457, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31882119

ABSTRACT

The transcription factor, MafB, plays important role in the differentiation and functional maintenance of various cells and tissues, such as the inner ear, kidney podocyte, parathyroid gland, pancreatic islet, and macrophages. The rare heterozygous substitution (p.Leu239Pro) of the DNA binding domain in MAFB is the cause of Focal Segmental Glomerulosclerosis associated with Duane Retraction Syndrome, which is characterized by impaired horizontal eye movement due to cranial nerve maldevelopment in humans. In this research, we generated mice carrying MafB p.Leu239Pro (Mafbmt/mt) and retrieved their tissues for analysis. As a result, we found that the phenotype of Mafbmt/mt mouse was similar to that of the conventional Mafb deficient mouse. This finding suggests that the Leucine residue at 239 in the DNA binding domain plays a key role in MafB function and could contribute to the diagnosis or development of treatment for patients carrying the MafB p.Leu239Pro missense variant.


Subject(s)
Ear/pathology , Kidney/pathology , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism , Mutation , Animals , Animals, Newborn , Binding Sites , DNA/metabolism , Ear/embryology , Humans , Kidney/embryology , Mice, Knockout , Mice, Mutant Strains , Mutation, Missense , Pancreas/pathology , Parathyroid Hormone/metabolism , Phenotype
7.
Exp Anim ; 69(1): 1-10, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31582643

ABSTRACT

The transcription factor MafB regulates macrophage differentiation. However, studies on the phenotype of Mafb-deficient macrophages are still limited. Recently, it was shown that the specific expression of MafB permits macrophages to be distinguished from dendritic cells. In addition, MafB has been reported to be involved in various diseases related to macrophages. Studies using macrophage-specific Mafb-deficient mice show that MafB is linked to atherosclerosis, autoimmunity, obesity, and ischemic stroke, all of which exhibit macrophage abnormality. Therefore, MafB is hypothesized to be indispensable for the regulation of macrophages to maintain systemic homeostasis and may serve as an innovative target for treating macrophage-related diseases.


Subject(s)
Macrophages/metabolism , MafB Transcription Factor/genetics , Atherosclerosis/metabolism , Autoimmunity , Homeostasis , MafB Transcription Factor/metabolism , Obesity/metabolism , Stroke/metabolism
8.
Biochem Biophys Res Commun ; 521(3): 590-595, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31679694

ABSTRACT

The transcription factor MafB is specifically expressed in macrophages. We have recently demonstrated that MafB is expressed in anti-inflammatory alternatively activated M2 macrophages in vitro. Tumor-associated macrophages (TAMs) are a subset of M2 type macrophages that can promote immunosuppressive activity, induce angiogenesis, and promote tumor cell proliferation. To examine whether MafB express in TAMs, we analyzed green fluorescent protein (GFP) expression in Lewis lung carcinoma tumors of MafB-GFP knock-in heterozygous mice. FACS analysis demonstrated GFP fluorescence in cells positive for macrophage-markers (F4/80, CD11b, CD68, and CD204). Moreover, quantitative RT-PCR analysis with F4/80+GFP+ and F4/80+GFP- sorted cells showed that the GFP-positive macrophages express IL-10, Arg-1, and TNF-α, which were known to be expressed in TAMs. These results indicate that MafB is expressed in TAMs. Furthermore, immunostaining analysis using an anti-MAFB antibody revealed that MAFB is expressed in CD204-and CD68-positive macrophages in human lung cancer samples. In conclusion, MafB can be a suitable marker of TAMs in both mouse and human tumor tissues.


Subject(s)
Carcinoma, Lewis Lung/pathology , Lung Neoplasms/pathology , Macrophages/pathology , MafB Transcription Factor/analysis , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Tumor Microenvironment
9.
Sci Rep ; 9(1): 19866, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882694

ABSTRACT

The environment experienced during spaceflight may impact the immune system and the thymus appears to undergo atrophy during spaceflight. However, molecular aspects of this thymic atrophy remain to be elucidated. In this study, we analysed the thymi of mice on board the international space station (ISS) for approximately 1 month. Thymic size was significantly reduced after spaceflight. Notably, exposure of mice to 1 × g using centrifugation cages in the ISS significantly mitigated the reduction in thymic size. Although spaceflight caused thymic atrophy, the global thymic structure was not largely changed. However, RNA sequencing analysis of the thymus showed significantly reduced expression of cell cycle-regulating genes in two independent spaceflight samples. These reductions were partially countered by 1 × g exposure during the space flights. Thus, our data suggest that spaceflight leads to reduced proliferation of thymic cells, thereby reducing the size of the thymus, and exposure to 1 × g might alleviate the impairment of thymus homeostasis induced by spaceflight.


Subject(s)
Gravity, Altered , Space Flight , Thymus Gland/metabolism , Animals , Base Sequence , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred C57BL , Principal Component Analysis , RNA-Seq
10.
Sci Rep ; 9(1): 7654, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31114014

ABSTRACT

Secondary lymphoid organs are critical for regulating acquired immune responses. The aim of this study was to characterize the impact of spaceflight on secondary lymphoid organs at the molecular level. We analysed the spleens and lymph nodes from mice flown aboard the International Space Station (ISS) in orbit for 35 days, as part of a Japan Aerospace Exploration Agency mission. During flight, half of the mice were exposed to 1 g by centrifuging in the ISS, to provide information regarding the effect of microgravity and 1 g exposure during spaceflight. Whole-transcript cDNA sequencing (RNA-Seq) analysis of the spleen suggested that erythrocyte-related genes regulated by the transcription factor GATA1 were significantly down-regulated in ISS-flown vs. ground control mice. GATA1 and Tal1 (regulators of erythropoiesis) mRNA expression was consistently reduced by approximately half. These reductions were not completely alleviated by 1 g exposure in the ISS, suggesting that the combined effect of space environments aside from microgravity could down-regulate gene expression in the spleen. Additionally, plasma immunoglobulin concentrations were slightly altered in ISS-flown mice. Overall, our data suggest that spaceflight might disturb the homeostatic gene expression of the spleen through a combination of microgravity and other environmental changes.


Subject(s)
GATA1 Transcription Factor/metabolism , Space Flight , Spleen/metabolism , Transcriptome , Animals , Down-Regulation , Erythropoiesis , GATA1 Transcription Factor/genetics , Mice , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Weightlessness/adverse effects
11.
Exp Anim ; 68(1): 103-111, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30369533

ABSTRACT

Multicentric carpotarsal osteolysis (MCTO) is a condition involving progressive osteolysis of the carpal and tarsal bones that is associated with glomerular sclerosis and renal failure (MCTO nephropathy). Previous work identified an autosomal dominant missense mutation in the transactivation domain of the transcription factor MAFB as the cause of MCTO. Several methods are currently used for MCTO nephropathy treatment, but these methods are invasive and lead to severe side effects, limiting their use. Therefore, the development of alternative treatments for MCTO nephropathy is required; however, the pathogenesis of MCTO in vivo is unclear without access to a mouse model. Here, we report the generation of an MCTO mouse model using the CRISPR/Cas9 system. These mice exhibit nephropathy symptoms that are similar to those observed in MCTO patients. MafbMCTO/MCTO mice show developmental defects in body weight from postnatal day 0, which persist as they age. They also exhibit high urine albumin creatinine levels from a young age, mimicking the nephropathic symptoms of MCTO patients. Characteristics of glomerular sclerosis reported in human patients are also observed, such as histological evidence of focal segmental glomerulosclerosis (FSGS), podocyte foot process microvillus transformation and podocyte foot process effacement. Therefore, this study contributes to the development of an alternative treatment for MCTO nephropathy by providing a viable mouse model.


Subject(s)
Disease Models, Animal , Glomerulosclerosis, Focal Segmental/genetics , MafB Transcription Factor/genetics , Mutation, Missense/genetics , Osteolysis/genetics , Renal Insufficiency/genetics , Albuminuria , Animals , Body Weight/genetics , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , Creatinine/urine , Glomerulosclerosis, Focal Segmental/therapy , Humans , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Mutant Strains , Osteolysis/therapy , Renal Insufficiency/therapy , Transcriptional Activation/genetics
12.
PLoS One ; 13(11): e0207321, 2018.
Article in English | MEDLINE | ID: mdl-30452437

ABSTRACT

Mouse embryonic stem cells (ESCs) are pluripotent stem cells, which have the ability to differentiate into all three germ layers: mesoderm, endoderm, and ectoderm. Proper levels of phosphorylated extracellular signal-regulated kinase (pERK) are critical for maintaining pluripotency, as elevated pERK evoked by fibroblast growth factor (FGF) receptor activation results in differentiation of ESCs, while, conversely, reduction of pERK by a MEK inhibitor maintains a pluripotent ground state. However, mechanisms underlying proper control of pERK levels in mouse ESCs are not fully understood. Here, we find that Klf5, a Krüppel-like transcription factor family member, is a component of pERK regulation in mouse ESCs. We show that ERK signaling is overactivated in Klf5-KO ESCs and the overactivated ERK in Klf5-KO ESCs is suppressed by the introduction of Klf5, but not Klf2 or Klf4, indicating a unique role for Klf5 in ERK suppression. Moreover, Klf5 regulates Spred1, a negative regulator of the FGF-ERK pathway. Klf5 also facilitates reprogramming of EpiSCs into a naïve state in combination with a glycogen synthase kinase 3 inhibitor and LIF, and in place of a MEK inhibitor. Taken together, these results show for the first time that Klf5 has a unique role suppressing ERK activity in mouse ESCs.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , MAP Kinase Signaling System , Mouse Embryonic Stem Cells/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Repressor Proteins/genetics , Repressor Proteins/metabolism
13.
Biochem Biophys Res Commun ; 501(3): 745-750, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29753741

ABSTRACT

Hindlimb unloading (HU) of rodents has been used as a ground-based model of spaceflight. In this study, we investigated the detailed impact of 14-day HU on the murine thymus. Thymic mass and cell number were significantly reduced after 14 days of hindlimb unloading, which was accompanied by an increment of plasma corticosterone. Although corticosterone reportedly causes selective apoptosis of CD4+CD8+ thymocytes (CD4+CD8+DPs) in mice treated with short-term HU, the reduction of thymocyte cellularity after the 14-day HU was not selective for CD4+CD8+DPs. In addition to the thymocyte reduction, the cellularity of thymic epithelial cells (TECs) was also reduced by the 14-day HU. Flow cytometric and RNA-sequencing analysis suggested that medullary TECs (mTECs) were preferentially reduced after HU. Moreover, immunohistochemical staining suggested that the 14-day HU caused a reduction of the mTECs expressing autoimmune regulator (Aire). Our data suggested that HU impacts both thymocytes and TECs. Consequently, these data imply that thymic T cell repertoire formation could be disturbed during spaceflight-like stress.


Subject(s)
Epithelial Cells/cytology , Hindlimb Suspension/methods , Thymocytes/cytology , Thymus Gland/physiology , Transcription Factors/analysis , Animals , CD4 Antigens/analysis , CD8 Antigens/analysis , Cell Count , Male , Mice, Inbred C57BL , Organ Size , Thymus Gland/cytology , Time Factors , AIRE Protein
14.
Nat Commun ; 8(1): 1700, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167450

ABSTRACT

The transcription factor MafB is expressed by monocytes and macrophages. Efferocytosis (apoptotic cell uptake) by macrophages is important for inhibiting the development of autoimmune diseases, and is greatly reduced in Mafb-deficient macrophages. Here, we show the expression of the first protein in the classical complement pathway C1q is important for mediating efferocytosis and is reduced in Mafb-deficient macrophages. The efferocytosis defect in Mafb-deficient macrophages can be rescued by adding serum from wild-type mice, but not by adding serum from C1q-deficient mice. By hemolysis assay we also show that activation of the classical complement pathway is decreased in Mafb-deficient mice. In addition, MafB overexpression induces C1q-dependent gene expression and signals that induce C1q genes are less effective in the absence of MafB. We also show that Mafb-deficiency can increase glomerular autoimmunity, including anti-nuclear antibody deposition. These results show that MafB is an important regulator of C1q.


Subject(s)
Complement C1q/metabolism , MafB Transcription Factor/immunology , Animals , Apoptosis/immunology , Autoimmunity , Complement C1q/deficiency , Complement C1q/genetics , Complement Pathway, Classical , Gene Expression Regulation , Gene Knockout Techniques , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , MafB Transcription Factor/deficiency , MafB Transcription Factor/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , RAW 264.7 Cells , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
15.
Development ; 144(20): 3706-3718, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28870993

ABSTRACT

The inner cell mass of the mouse blastocyst gives rise to the pluripotent epiblast (EPI), which forms the embryo proper, and the primitive endoderm (PrE), which forms extra-embryonic yolk sac tissues. All inner cells coexpress lineage markers such as Nanog and Gata6 at embryonic day (E) 3.25, and the EPI and PrE precursor cells eventually segregate to exclusively express Nanog and Gata6, respectively. Fibroblast growth factor (FGF)-extracellular signal-regulated kinase (ERK) signalling is involved in segregation of the EPI and PrE lineages; however, the mechanism involved in Fgf4 regulation is poorly understood. Here, we identified Klf5 as an upstream repressor of Fgf4Fgf4 was markedly upregulated in Klf5 knockout (KO) embryos at E3.0, and was downregulated in embryos overexpressing Klf5 Furthermore, Klf5 KO and overexpressing blastocysts showed skewed lineage specification phenotypes, similar to FGF4-treated preimplantation embryos and Fgf4 KO embryos, respectively. Inhibitors of the FGF receptor (Fgfr) and ERK pathways reversed the skewed lineage specification of Klf5 KO blastocysts. These data demonstrate that Klf5 suppresses Fgf4-Fgfr-ERK signalling, thus preventing precocious activation of the PrE specification programme.


Subject(s)
Endoderm/metabolism , Fibroblast Growth Factor 4/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/metabolism , Animals , Blastocyst/metabolism , Cell Differentiation , Cell Lineage , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Pluripotent Stem Cells/cytology , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Time Factors
16.
FEBS Open Bio ; 6(6): 540-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27419056

ABSTRACT

MafB, a transcription factor expressed selectively in macrophages, has important roles in some macrophage-related diseases, especially in atherosclerosis. In this study, we investigated the mechanism by which hematopoietic-specific MafB deficiency induces the development of obesity. Wild-type and hematopoietic cell-specific Mafb-deficient mice were fed a high-fat diet for 10 weeks. The Mafb-deficient mice exhibited higher body weights and faster rates of body weight increase than control mice. The Mafb-deficient mice also had a higher percentage of body fat than the wild-type mice, due to increased adipocyte size and serum cholesterol levels. Reverse transcription-PCR analysis showed a reduction in apoptosis inhibitor of macrophage (AIM) in Mafb-deficient adipose tissue. AIM is known as an inhibitor of lipogenesis in adipocytes and is expressed in adipose tissue macrophages. Collectively, our data suggest that Mafb deficiency in hematopoietic cells accelerates the development of obesity.

17.
Biochem Biophys Res Commun ; 473(1): 118-124, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26996125

ABSTRACT

The large Maf transcription factors c-Maf and MafB are expressed in macrophage-lineage hematopoietic cells, but the expression patterns of MafB and c-Maf in macrophage subtypes and tissue-resident macrophages have not been fully analyzed. First, we analyzed MafB and c-Maf protein expression in tissue-resident macrophages. Mouse lymph nodes, spleens, lungs, and kidneys were subjected to immunohistochemistry using anti-MafB and anti-c-Maf. Both MafB and c-Maf signals were observed in lymph node macrophages. In the splenic macrophages the MafB signal was detected by anti-MafB, but the c-Maf signal was not detected. No expression of c-Maf or MafB was detected in macrophages in the lung and kidney. Flow cytometry analysis revealed a similar pattern of GFP expression in Mafb/GFP knock-in heterozygous mice. To analyze these different expression patterns in greater detail, we examined the expression of MafB and c-Maf by quantitative RT-PCR in different cytokine- or LPS-induced macrophages in vitro. MafB expression was induced by IL-10 or IL-4 with IL-13 and was reduced by LPS or GM-CSF. By contrast, c-Maf expression was induced by IL-10 and reduced by IL-4 with IL-13 or GM-CSF. These results indicate that MafB and c-Maf have different expression patterns in macrophages, suggesting differences in function.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , MafB Transcription Factor/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Animals , Bronchoalveolar Lavage , Cell Separation , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Green Fluorescent Proteins/metabolism , Heterozygote , Interleukin-10/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Kidney/metabolism , Lipopolysaccharides/chemistry , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Signal Transduction , Tissue Distribution
18.
PLoS One ; 11(3): e0150715, 2016.
Article in English | MEDLINE | ID: mdl-26943822

ABSTRACT

Pluripotency is maintained in mouse embryonic stem (ES) cells and is induced from somatic cells by the activation of appropriate transcriptional regulatory networks. Krüppel-like factor gene family members, such as Klf2, Klf4 and Klf5, have important roles in maintaining the undifferentiated state of mouse ES cells as well as in cellular reprogramming, yet it is not known whether other Klf family members exert self-renewal and reprogramming functions when overexpressed. In this study, we examined whether overexpression of any representative Klf family member, such as Klf1-Klf10, would be sufficient for the self-renewal of mouse ES cells. We found that only Klf2, Klf4, and Klf5 produced leukemia inhibitory factor (LIF)-independent self-renewal, although most KLF proteins, if not all, have the ability to occupy the regulatory regions of Nanog, a critical Klf target gene. We also examined whether overexpression of any of Klf1-Klf10 would be sufficient to convert epiblast stem cells into a naïve pluripotent state and found that Klf5 had such reprogramming ability, in addition to Klf2 and Klf4. We also delineated the functional domains of the Klf2 protein for LIF-independent self-renewal and reprogramming. Interestingly, we found that both the N-terminal transcriptional activation and C-terminal zinc finger domains were indispensable for this activity. Taken together, our comprehensive analysis provides new insight into the contribution of Klf family members to mouse ES self-renewal and cellular reprogramming.


Subject(s)
Cell Self Renewal , Cellular Reprogramming , Kruppel-Like Transcription Factors/metabolism , Mouse Embryonic Stem Cells/cytology , Multigene Family , Animals , Chimera , Chromatin Immunoprecipitation , Epitopes/metabolism , Germ Layers/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/chemistry , Leukemia Inhibitory Factor/metabolism , Mice , Protein Structure, Tertiary
19.
Exp Anim ; 65(2): 175-87, 2016 May 20.
Article in English | MEDLINE | ID: mdl-26822934

ABSTRACT

The Japan Aerospace Exploration Agency developed the mouse Habitat Cage Unit (HCU) for installation in the Cell Biology Experiment Facility (CBEF) onboard the Japanese Experimental Module ("Kibo") on the International Space Station. The CBEF provides "space-based controls" by generating artificial gravity in the HCU through a centrifuge, enabling a comparison of the biological consequences of microgravity and artificial gravity of 1 g on mice housed in space. Therefore, prior to the space experiment, a ground-based study to validate the habitability of the HCU is necessary to conduct space experiments using the HCU in the CBEF. Here, we investigated the ground-based effect of a 32-day housing period in the HCU breadboard model on male mice in comparison with the control cage mice. Morphology of skeletal muscle, the thymus, heart, and kidney, and the sperm function showed no critical abnormalities between the control mice and HCU mice. Slight but significant changes caused by the HCU itself were observed, including decreased body weight, increased weights of the thymus and gastrocnemius, reduced thickness of cortical bone of the femur, and several gene expressions from 11 tissues. Results suggest that the HCU provides acceptable conditions for mouse phenotypic analysis using CBEF in space, as long as its characteristic features are considered. Thus, the HCU is a feasible device for future space experiments.


Subject(s)
Gravitation , Housing, Animal , Phenotype , Space Flight , Weightlessness , Animals , Femur/anatomy & histology , Heart/anatomy & histology , Kidney/anatomy & histology , Male , Mice, Inbred C57BL , Muscle, Skeletal/anatomy & histology , Spermatozoa/physiology , Thymus Gland/anatomy & histology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...