Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Sci Rep ; 8(1): 14515, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30266983

ABSTRACT

By using near-infrared fluorescent protein (iRFP)-expressing hematopoietic cells, we established a novel, quantitative, in vivo, noninvasive atherosclerosis imaging system. This murine atherosclerosis imaging approach targets macrophages expressing iRFP in plaques. Low-density lipoprotein receptor-deficient (LDLR-/-) mice transplanted with beta-actin promoter-derived iRFP transgenic (TG) mouse bone marrow (BM) cells (iRFP → LDLR-/-) were used. Atherosclerosis was induced by a nonfluorescent 1.25% cholesterol diet (HCD). Atherosclerosis was compared among the three differently induced mouse groups. iRFP → LDLR-/- mice fed a normal diet (ND) and LDLR-/- mice transplanted with wild-type (WT) BM cells were used as controls. The in vivo imaging system (IVIS) detected an enhanced iRFP signal in the thoracic aorta of HCD-fed iRFP → LDLR-/- mice, whereas iRFP signals were not observed in the control mice. Time-course imaging showed a gradual increase in the signal area, which was correlated with atherosclerotic plaque progression. Oil red O (ORO) staining of aortas and histological analysis of plaques confirmed that the detected signal was strictly emitted from plaque-positive areas of the aorta. Our new murine atherosclerosis imaging system can noninvasively image atherosclerotic plaques in the aorta and generate longitudinal data, validating the ability of the system to monitor lesion progression.


Subject(s)
Aortic Diseases/diagnostic imaging , Atherosclerosis/diagnostic imaging , Luminescent Measurements/methods , Luminescent Proteins/analysis , Optical Imaging/methods , Plaque, Atherosclerotic/diagnostic imaging , Actins/genetics , Animals , Aortic Diseases/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Azo Compounds , Bone Marrow Transplantation , Cholesterol, Dietary/toxicity , Coloring Agents , Flow Cytometry , Genes, Reporter , Genes, Synthetic , Luminescent Proteins/genetics , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/ultrastructure , Mice , Mice, Transgenic , Microscopy, Fluorescence , Promoter Regions, Genetic , Receptors, LDL/deficiency , Recombinant Proteins/analysis , Recombinant Proteins/genetics
3.
Kidney Int ; 94(2): 396-407, 2018 08.
Article in English | MEDLINE | ID: mdl-29779709

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a leading cause of end-stage renal disease in children and adults. Genetic factors significantly contribute to early-onset FSGS, but the etiologies of most adult cases remain unknown. Genetic studies of monogenic syndromic FSGS exhibiting extra-renal manifestations have uncovered an unexpected biological role for genes in the development of both podocytes and other cellular lineages. To help define these roles, we studied two unrelated families with FSGS associated with Duane Retraction Syndrome, characterized by impaired horizontal eye movement due to cranial nerve malformation. All four affected individuals developed FSGS and Duane Retraction Syndrome in their first to second decade of life, manifested as restricted abduction together with globe retraction and narrowed palpebral fissure on attempted adduction. Hypoplasia of the abducens nerves and hearing impairment occurred in severely affected individuals. Genetic analyses revealed that affected individuals harbor a rare heterozygous substitution (p.Leu239Pro) in MAFB, a leucine zipper transcription factor. Luciferase assays with cultured monocytes indicated that the substitution significantly reduced transactivation of the F4/80 promoter, the known MAFB recognition element. Additionally, immunohistochemistry indicated reduced MAFB expression in the podocytes of patients. Structural modeling suggested that the p.Leu239Pro substitution in the DNA-binding domain possibly interferes with the stability of the adjacent zinc finger. Lastly, podocytes in neonatal mice with p.Leu239Pro displayed impaired differentiation. Thus, MAFB mutations impair development and/or maintenance of podocytes, abducens neurons and the inner ear. The interactions between MAFB and regulatory elements in these developing organs are likely highly specific based on spatiotemporal requirements.


Subject(s)
Duane Retraction Syndrome/etiology , Glomerulosclerosis, Focal Segmental/genetics , Kidney Failure, Chronic/etiology , MafB Transcription Factor/genetics , Adolescent , Adult , Age of Onset , Amino Acid Substitution , Animals , Child , Duane Retraction Syndrome/pathology , Female , Genetic Testing , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/pathology , Heterozygote , Humans , Kidney Failure, Chronic/pathology , Male , Mice , Mutation , Podocytes/pathology , Protein Domains/genetics , Sequence Homology, Amino Acid , Young Adult
4.
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
5.
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.

6.
Exp Anim ; 63(3): 311-9, 2014.
Article in English | MEDLINE | ID: mdl-25077761

ABSTRACT

Fluorescent proteins with light wavelengths within the optical window are one of the improvements in in vivo imaging techniques. Near-infrared (NIR) fluorescent protein (iRFP) is a stable, nontoxic protein that emits fluorescence within the NIR optical window without the addition of exogenous substrate. However, studies utilizing an in vivo iRFP model have not yet been published. Here, we report the generation of transgenic iRFP mice with ubiquitous NIR fluorescence expression. iRFP expression was observed in approximately 50% of the offspring from a matings between iRFP transgenic and WT mice. The serum and blood cell indices and body weights of iRFP mice were similar to those of WT mice. Red fluorescence with an excitation wavelength of 690 nm and an emission wavelength of 713 nm was detected in both newborn and adult iRFP mice. We also detected fluorescence emission in whole organs of the iRFP mice, including the brain, heart, liver, kidney, spleen, lung, pancreas, bone, testis, thymus, and adipose tissue. Therefore, iRFP transgenic mice may therefore be a useful tool for various types of in vivo imaging.


Subject(s)
Luminescent Proteins/analysis , Mice, Transgenic , Molecular Imaging/methods , Spectrum Analysis/methods , Whole Body Imaging/methods , Animals
7.
Nat Commun ; 5: 3147, 2014.
Article in English | MEDLINE | ID: mdl-24445679

ABSTRACT

MafB is a transcription factor that induces myelomonocytic differentiation. However, the precise role of MafB in the pathogenic function of macrophages has never been clarified. Here we demonstrate that MafB promotes hyperlipidemic atherosclerosis by suppressing foam-cell apoptosis. Our data show that MafB is predominantly expressed in foam cells found within atherosclerotic lesions, where MafB mediates the oxidized LDL-activated LXR/RXR-induced expression of apoptosis inhibitor of macrophages (AIM). In the absence of MafB, activated LXR/RXR fails to induce the expression of AIM, a protein that is normally responsible for protecting macrophages from apoptosis; thus, Mafb-deficient macrophages are prone to apoptosis. Haematopoietic reconstitution with Mafb-deficient fetal liver cells in recipient LDL receptor-deficient hyperlipidemic mice revealed accelerated foam-cell apoptosis, which subsequently led to the attenuation of the early atherogenic lesion. These findings represent the first evidence that the macrophage-affiliated MafB transcription factor participates in the acceleration of atherogenesis.


Subject(s)
Apoptosis , Atherosclerosis/physiopathology , Foam Cells/pathology , MafB Transcription Factor/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Atherosclerosis/pathology , Base Sequence , Humans , Liver X Receptors , MafB Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Orphan Nuclear Receptors/metabolism , Receptors, Immunologic/genetics , Receptors, Scavenger , Retinoid X Receptors/metabolism , Sequence Homology, Nucleic Acid
8.
Blood ; 118(5): 1374-85, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21628412

ABSTRACT

c-Maf is one of the large Maf (musculoaponeurotic fibrosarcoma) transcription factors that belong to the activated protein-1 super family of basic leucine zipper proteins. Despite its overexpression in hematologic malignancies, the physiologic roles c-Maf plays in normal hematopoiesis have been largely unexplored. On a C57BL/6J background, c-Maf(-/-) embryos succumbed from severe erythropenia between embryonic day (E) 15 and E18. Flow cytometric analysis of fetal liver cells showed that the mature erythroid compartments were significantly reduced in c-Maf(-/-) embryos compared with c-Maf(+/+) littermates. Interestingly, the CFU assay indicated there was no significant difference between c-Maf(+/+) and c-Maf(-/-) fetal liver cells in erythroid colony counts. This result indicated that impaired definitive erythropoiesis in c-Maf(-/-) embryos is because of a non-cell-autonomous effect, suggesting a defective erythropoietic microenvironment in the fetal liver. As expected, the number of erythroblasts surrounding the macrophages in erythroblastic islands was significantly reduced in c-Maf(-/-) embryos. Moreover, decreased expression of VCAM-1 was observed in c-Maf(-/-) fetal liver macrophages. In conclusion, these results strongly suggest that c-Maf is crucial for definitive erythropoiesis in fetal liver, playing an important role in macrophages that constitute erythroblastic islands.


Subject(s)
Erythroblasts/cytology , Erythroblasts/physiology , Erythropoiesis/genetics , Fetus/cytology , Liver/cytology , Proto-Oncogene Proteins c-maf/physiology , Animals , Cell Communication/genetics , Cell Movement/genetics , Cell Proliferation , Embryo, Mammalian , Erythroblasts/metabolism , Fetus/metabolism , Gene Expression Profiling , Liver/embryology , Liver/metabolism , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Proto-Oncogene Proteins c-maf/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...