Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Enzymol ; 537: 123-39, 2014.
Article in English | MEDLINE | ID: mdl-24480344

ABSTRACT

Adipocytes reside in discrete, well-defined depots throughout the body. In addition to mature adipocytes, white adipose tissue depots are composed of many cell types, including macrophages, endothelial cells, fibroblasts, and stromal cells, which together are referred to as the stromal vascular fraction (SVF). The SVF also contains adipocyte progenitors that give rise to mature adipocytes in those depots. Marrow adipose tissue (MAT) or marrow fat has long been known to be present in bone marrow (BM) but its origin, development, and function remain largely unknown. Clinically, increased MAT is associated with age, metabolic diseases, drug treatment, and marrow recovery in children receiving radiation and chemotherapy. In contrast to the other depots, MAT is unevenly distributed in the BM of long bones. Conventional quantitation relies on sectioning of the bone to overcome issues with distribution but is time-consuming, resource intensive, inconsistent between laboratories and may be unreliable as it may miss changes in MAT volume. Thus, the inability to quantitate MAT in a rapid, systematic, and reproducible manner has hampered a full understanding of its development and function. In this chapter, we describe a new technique that couples histochemical staining of lipid using osmium tetroxide with microcomputerized tomography to visualize and quantitate MAT within the medullary canal in three dimensions. Imaging of osmium staining provides a high-resolution map of existing and developing MAT in the BM. Because this method is simple, reproducible, and quantitative, we expect it will become a useful tool for the precise characterization of MAT.


Subject(s)
Cell Differentiation , Osmium Tetroxide/chemistry , Staining and Labeling/methods , X-Ray Microtomography/methods , Adipogenesis/genetics , Adipose Tissue, White/growth & development , Bone Marrow/growth & development , Humans , Stromal Cells/cytology
2.
Kidney Int ; 85(5): 1091-102, 2014 May.
Article in English | MEDLINE | ID: mdl-24172684

ABSTRACT

The coordination of multiple cytokines and transcription factors with their downstream signaling pathways has been shown to be integral to nephron maturation. Here we present a completely novel role for the helix-loop-helix transcription factor Early B-cell factor 1 (Ebf1), originally identified for B-cell maturation, for the proper maturation of glomerular cells from mesenchymal progenitors. The expression of Ebf1 was both spatially and temporally regulated within the developing cortex and glomeruli. Using Ebf1-null mice, we then identified biochemical, metabolic, and histological abnormalities in renal development that arose in the absence of this transcription factor. In the Ebf1 knockout mice, the developed kidneys show thinned cortices and reduced glomerular maturation. The glomeruli showed abnormal vascularization and severely effaced podocytes. The mice exhibited early albuminuria and elevated blood urea nitrogen levels. Moreover, the glomerular filtration rate was reduced >66% and the expression of podocyte-derived vascular endothelial growth factor A was decreased compared with wild-type control mice. Thus, Ebf1 has a significant and novel role in glomerular development, podocyte maturation, and the maintenance of kidney integrity and function.


Subject(s)
Kidney Glomerulus/metabolism , Trans-Activators/metabolism , Age Factors , Albuminuria/genetics , Albuminuria/metabolism , Albuminuria/physiopathology , Animals , Blood Urea Nitrogen , Cell Differentiation , Cells, Cultured , Gene Expression Regulation, Developmental , Genotype , Glomerular Filtration Rate , Kidney Glomerulus/growth & development , Kidney Glomerulus/physiopathology , Kidney Glomerulus/ultrastructure , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organogenesis , Phenotype , Podocytes/metabolism , Podocytes/ultrastructure , Signal Transduction , Time Factors , Trans-Activators/deficiency , Trans-Activators/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
3.
Endocrinology ; 151(4): 1611-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20172967

ABSTRACT

We previously reported that mice deficient for the transcription factor early B-cell factor (Ebf1) exhibit markedly increased numbers of osteoblasts, bone formation rate, and serum osteocalcin, but the bone marrow of Ebf1(-/-) mice is also striking in its increased marrow adiposity. The purpose of this work was to analyze the metabolic phenotype that accompanies the altered bone morphology of Ebf1(-/-) mice. Whereas marrow adiposity was increased, deposition of white adipose tissue in other regions of the body was severely reduced (sc 40-50%, abdominally 80-85%). Brown adipose exhibited decreased lipid deposition. Subcutaneous and perigonadal white adipose tissue showed a decrease in mRNA transcripts for peroxisomal proliferator-activated receptor-gamma2 and CCAAT/enhancer-binding protein-beta in Ebf1(-/-) tissue compared with wild type. Circulating levels of leptin were decreased in Ebf1(-/-) animals compared with their littermate controls (down 65-95%), whereas adiponectin remained comparable after 2 wk of age. Serum analysis also found the Ebf1(-/-) animals were hypoglycemic and hypotriglyceridemic. After ip injection of insulin, the serum glucose levels in Ebf1(-/-) mice took longer to recover, and after a glucose challenge the Ebf1(-/-) animals reached serum glucose levels almost twice that of their wild-type counterparts. Measurement of circulating pancreatic hormones revealed normal or reduced insulin levels in the Ebf1(-/-) mice, whereas glucagon was significantly increased (up 1.7- to 8.5-fold). Metabolically the Ebf1(-/-) mice had increased O(2) consumption, CO(2) production, food and water intake, and activity. Markers for gluconeogenesis, however, were decreased in the Ebf1(-/-) mice compared with controls. In conclusion, the Ebf1-deficient animals exhibit defects in adipose tissue deposition with increased marrow adiposity and impaired glucose mobilization.


Subject(s)
Adipose Tissue/metabolism , Energy Metabolism/physiology , Lipid Metabolism/physiology , Lipodystrophy/metabolism , Trans-Activators/metabolism , Adiponectin/blood , Animals , Blood Glucose , Bone Marrow/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calorimetry , Glucagon/blood , Insulin/blood , Leptin/blood , Lipodystrophy/genetics , Mice , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Trans-Activators/genetics
4.
Bone ; 44(4): 537-46, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19130908

ABSTRACT

Ebf1 is a transcription factor essential for B cell fate specification and function and important for the development of olfactory sensory neurons. We show here that Ebf1 also plays an important role in regulating osteoblast and adipocyte development in vivo. Ebf1 mRNA and protein is expressed in MSCs, in OBs at most stages of differentiation, and in adipocytes. Tibiae and femora from Ebf1(-/-) mice had a striking increase in all bone formation parameters examined including the number of OBs, osteoid volume, and bone formation rate. Serum osteocalcin, a marker of bone formation, was significantly elevated in mutant mice. The numbers of osteoclasts in bone were normal in younger (4 week-old) Ebf1(-/-) mice but increased in older (12 week-old) Ebf1(-/-) mice. This correlated well with in vitro osteoclast development from bone marrow cells. In addition to the increased osteoblastogenesis, there was a dramatic increase in adipocyte numbers in the bone marrow of Ebf1(-/-) mice. Increased adiposity was also seen histologically in the liver but not in the spleen of these mice, and accompanied by decreased deposition of adipose to subcutaneous sites. Thus Ebf1-deficient mice appear to be a new model of lipodystrophy. Ebf1 is a rare example of a transcription factor that regulates both the osteoblast and adipocyte lineages similarly.


Subject(s)
Adipocytes/cytology , Cell Differentiation/physiology , Osteoblasts/cytology , Osteogenesis/physiology , Trans-Activators/metabolism , Adipocytes/metabolism , Animals , Blotting, Western , Cell Lineage , Immunohistochemistry , Mice , Mice, Knockout , Osteoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
5.
J Immunol ; 173(11): 6583-91, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15557148

ABSTRACT

Pax5 encodes BSAP, a member of the paired box domain transcription factors, whose expression is restricted to B lymphocyte lineage cells. Pax5(-/-) mice have a developmental arrest of the B cell lineage at the pro-B cell stage. We show here that Pax5(-/-) mice are severely osteopenic, missing 60% of their bone mass. The osteopenia can be accounted for by a >100% increase in the number of osteoclasts in bone measured histomorphometrically. This is not due to a lack of B cells, because other strains of B cell-deficient mice do not exhibit this phenotype. There was no difference in the number of osteoclasts produced in vitro by wild-type and Pax5(-/-) bone marrow cells. In contrast, spleen cells from Pax5(-/-) mice produce as much as five times the number of osteoclasts as control spleen cells. Culture of Pax5(-/-) spleen cells yields a population of adherent cells that grow spontaneously in culture without added growth factors for >4 wk. These cells have a monocyte phenotype, produce large numbers of osteoclasts when induced in vitro, and therefore are highly enriched in osteoclast precursors. These data demonstrate a previously unsuspected connection between B cell and osteoclast development and a key role for Pax5 in the control of osteoclast development.


Subject(s)
Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Osteoclasts/pathology , Stem Cells/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Bone Diseases, Metabolic/immunology , Cell Count , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/biosynthesis , Growth Disorders/genetics , Growth Disorders/immunology , Growth Disorders/pathology , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , PAX5 Transcription Factor , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Stem Cells/metabolism , Transcription Factors/biosynthesis
6.
J Bone Miner Res ; 19(4): 652-60, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15005853

ABSTRACT

UNLABELLED: Mice deficient in GATA-1 or NF-E2 have a 200-300% increase in bone volume and formation parameters. Osteoblasts and osteoclasts generated in vitro from mutant and control animals were similar in number and function. Osteoblast proliferation increased up to 6-fold when cultured with megakaryocytes. A megakaryocyte-osteoblast interaction plays a role in the increased bone formation in these mice. INTRODUCTION: GATA-1 and NF-E2 are transcription factors required for the differentiation of megakaryocytes. Mice deficient in these factors have phenotypes characterized by markedly increased numbers of immature megakaryocytes, a concomitant drastic reduction of platelets, and a striking increased bone mass. The similar bone phenotype in both animal models led us to explore the interaction between osteoblasts and megakaryocytes. MATERIALS AND METHODS: Histomorphometry, microCT, and serum and urine biochemistries were used to assess the bone phenotype in these mice. Wildtype and mutant osteoblasts were examined for differences in proliferation, alkaline phosphatase activity, and osteocalcin secretion. In vitro osteoclast numbers and resorption were measured. Because mutant osteoblasts and osteoclasts were similar to control cells, and because of the similar bone phenotype, we explored the interaction between cells of the osteoblast lineage and megakaryocytes. RESULTS: A marked 2- to 3-fold increase in trabecular bone volume and bone formation indices were observed in these mice. A 20- to 150-fold increase in trabecular bone volume was measured for the entire femoral medullary canal. The increased bone mass phenotype in these animals was not caused by osteoclast defects, because osteoclast number and function were not compromised in vitro or in vivo. In contrast, in vivo osteoblast number and bone formation parameters were significantly elevated. When wildtype or mutant osteoblasts were cultured with megakaryocytes from GATA-1- or NF-E2-deficient mice, osteoblast proliferation increased over 3- to 6-fold by a mechanism that required cell-to-cell contact. CONCLUSIONS: These observations show an interaction between megakaryocytes and osteoblasts, which results in osteoblast proliferation and increased bone mass, and may represent heretofore unrecognized anabolic pathways in bone.


Subject(s)
Cell Communication/physiology , DNA-Binding Proteins/genetics , Megakaryocytes/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Transcription Factors/genetics , Alkaline Phosphatase/biosynthesis , Animals , Bone Density/genetics , Bone Development/genetics , Bone Development/physiology , Cell Communication/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/genetics , Cell Division/physiology , Cells, Cultured , DNA-Binding Proteins/deficiency , Erythroid-Specific DNA-Binding Factors , Femur/diagnostic imaging , Femur/physiopathology , GATA1 Transcription Factor , Megakaryocytes/cytology , Mice , Mice, Knockout , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Osteoblasts/cytology , Osteocalcin/biosynthesis , Osteoclasts/cytology , Radiography , Tibia/physiopathology , Tibia/ultrastructure , Transcription Factors/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...