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.
Mol Metab ; 20: 38-50, 2019 02.
Article in English | MEDLINE | ID: mdl-30553769

ABSTRACT

OBJECTIVE: Sympathetic nervous system and immune cell interactions play key roles in the regulation of metabolism. For example, recent convergent studies have shown that macrophages regulate obesity through brown adipose tissue (BAT) activation and beiging of white adipose tissue (WAT) via effects upon local catecholamine availability. However, these studies have raised issues about the underlying mechanisms involved including questions regarding the production of catecholamines by macrophages, the role of macrophage polarization state and the underlying intracellular signaling pathways in macrophages that might mediate these effects. METHODS: To address such issues we generated mice lacking Irs2, which mediates the effects of insulin and interleukin 4, specifically in LyzM expressing cells (Irs2LyzM-/- mice). RESULTS: These animals displayed obesity resistance and preservation of glucose homeostasis on high fat diet feeding due to increased energy expenditure via enhanced BAT activity and WAT beiging. Macrophages per se did not produce catecholamines but Irs2LyzM-/- mice displayed increased sympathetic nerve density and catecholamine availability in adipose tissue. Irs2-deficient macrophages displayed an anti-inflammatory transcriptional profile and alterations in genes involved in scavenging catecholamines and supporting increased sympathetic innervation. CONCLUSIONS: Our studies identify a critical macrophage signaling pathway involved in the regulation of adipose tissue sympathetic nerve function that, in turn, mediates key neuroimmune effects upon systemic metabolism. The insights gained may open therapeutic opportunities for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Insulin Receptor Substrate Proteins/genetics , Monocyte-Macrophage Precursor Cells/metabolism , Obesity/genetics , Sympathetic Nervous System/metabolism , Adipose Tissue, Brown/physiology , Animals , Catecholamines/metabolism , Cells, Cultured , Energy Metabolism , Gene Deletion , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Sympathetic Nervous System/physiology
2.
J Clin Invest ; 122(10): 3490-503, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945629

ABSTRACT

Serotonin is a critical regulator of bone mass, fulfilling different functions depending on its site of synthesis. Brain-derived serotonin promotes osteoblast proliferation, whereas duodenal-derived serotonin suppresses it. To understand the molecular mechanisms of duodenal-derived serotonin action on osteoblasts, we explored its transcriptional mediation in mice. We found that the transcription factor FOXO1 is a crucial determinant of the effects of duodenum-derived serotonin on bone formation We identified two key FOXO1 complexes in osteoblasts, one with the transcription factor cAMP-responsive element-binding protein 1 (CREB) and another with activating transcription factor 4 (ATF4). Under normal levels of circulating serotonin, the proliferative activity of FOXO1 was promoted by a balance between its interaction with CREB and ATF4. However, high circulating serotonin levels prevented the association of FOXO1 with CREB, resulting in suppressed osteoblast proliferation. These observations identify FOXO1 as the molecular node of an intricate transcriptional machinery that confers the signal of duodenal-derived serotonin to inhibit bone formation.


Subject(s)
Bone Remodeling/physiology , Duodenum/metabolism , Forkhead Transcription Factors/physiology , Osteoblasts/physiology , Serotonin/physiology , Activating Transcription Factor 4/physiology , Animals , Blood-Brain Barrier , Cell Division/drug effects , Cells, Cultured/drug effects , Cyclic AMP Response Element-Binding Protein/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genes, Reporter , Genotype , Homeostasis/physiology , Insulin-Like Growth Factor I/genetics , Low Density Lipoprotein Receptor-Related Protein-5/deficiency , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/physiology , MAP Kinase Signaling System/drug effects , Mice , Organ Specificity , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/physiology , Promoter Regions, Genetic , Receptor, Serotonin, 5-HT1B/deficiency , Receptor, Serotonin, 5-HT1B/genetics , Receptor, Serotonin, 5-HT1B/physiology , Serotonin/blood , Serotonin/pharmacology , Stress, Physiological/genetics , Transcription, Genetic
3.
J Biol Chem ; 287(12): 8757-68, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22298775

ABSTRACT

The Forkhead transcription factor FoxO1 inhibits through its expression in osteoblasts ß-cell proliferation, insulin secretion, and sensitivity. At least part of the FoxO1 metabolic functions result from its ability to suppress the activity of osteocalcin, an osteoblast-derived hormone favoring glucose metabolism and energy expenditure. In searching for mechanisms mediating the metabolic actions of FoxO1, we focused on ATF4, because this transcription factor also affects glucose metabolism through its expression in osteoblasts. We show here that FoxO1 co-localizes with ATF4 in the osteoblast nucleus, and physically interacts with and promotes the transcriptional activity of ATF4. Genetic experiments demonstrate that FoxO1 and ATF4 cooperate to increase glucose levels and decrease glucose tolerance. These effects result from a synergistic effect of the two transcription factors to suppress the activity of osteocalcin through up-regulating expression of the phosphatase catalyzing osteocalcin inactivation. As a result, insulin production by ß-cells and insulin signaling in the muscle, liver and white adipose tissue are compromised and fat weight increases by the FoxO1/ATF4 interaction. Taken together these observations demonstrate that FoxO1 and ATF4 cooperate in osteoblasts to regulate glucose homeostasis.


Subject(s)
Activating Transcription Factor 4/metabolism , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Osteoblasts/metabolism , Activating Transcription Factor 4/genetics , Animals , Cell Proliferation , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Homeostasis , Insulin/metabolism , Male , Mice , Mice, Knockout , Osteoblasts/cytology , Protein Binding
4.
Cell Metab ; 11(2): 147-60, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20142102

ABSTRACT

Osteoporosis, a disease of low bone mass, is associated with decreased osteoblast numbers and increased levels of oxidative stress within osteoblasts. Since transcription factors of the FoxO family confer stress resistance, we investigated their potential impact on skeletal integrity. Here we employ cell-specific deletion and molecular analyses to show that, among the three FoxO proteins, only FoxO1 is required for proliferation and redox balance in osteoblasts and thereby controls bone formation. FoxO1 regulation of osteoblast proliferation occurs through its interaction with ATF4, a transcription factor regulating amino acid import, as well as through its regulation of a stress-dependent pathway influencing p53 signaling. Accordingly, decreasing oxidative stress levels or increasing protein intake normalizes bone formation and bone mass in mice lacking FoxO1 specifically in osteoblasts. These results identify FoxO1 as a crucial regulator of osteoblast physiology and provide a direct mechanistic link between oxidative stress and the regulation of bone remodeling.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Osteoblasts/metabolism , Osteogenesis , Oxidative Stress , Activating Transcription Factor 4/metabolism , Animals , Cell Proliferation , Cells, Cultured , Forkhead Box Protein O1 , Gene Expression Regulation , Mice , Mutation , Protein Biosynthesis
5.
J Clin Invest ; 120(1): 357-68, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20038793

ABSTRACT

Osteoblasts have recently been found to play a role in regulating glucose metabolism through secretion of osteocalcin. It is unknown, however, how this osteoblast function is regulated transcriptionally. As FoxO1 is a forkhead family transcription factor known to regulate several key aspects of glucose homeostasis, we investigated whether its expression in osteoblasts may contribute to its metabolic functions. Here we show that mice lacking Foxo1 only in osteoblasts had increased pancreatic beta cell proliferation, insulin secretion, and insulin sensitivity. The ability of osteoblast-specific FoxO1 deficiency to affect metabolic homeostasis was due to increased osteocalcin expression and decreased expression of Esp, a gene that encodes a protein responsible for decreasing the bioactivity of osteocalcin. These results indicate that FoxO1 expression in osteoblasts contributes to FoxO1 control of glucose homeostasis and identify FoxO1 as a key modulator of the ability of the skeleton to function as an endocrine organ regulating glucose metabolism.


Subject(s)
Forkhead Transcription Factors/physiology , Glucose/metabolism , Homeostasis , Osteoblasts/physiology , Osteocalcin/physiology , Adenosine Triphosphate/biosynthesis , Adipokines/physiology , Animals , COS Cells , Cell Proliferation , Chlorocebus aethiops , Forkhead Box Protein O1 , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Mice , Obesity/prevention & control , Protein Tyrosine Phosphatases/physiology , Signal Transduction
6.
Hum Mol Genet ; 18(16): 3026-38, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19457928

ABSTRACT

The question of whether DNA methylation contributes to the stabilization of gene expression patterns in differentiated mammalian tissues remains controversial. Using genome-wide methylation profiling, we screened 3757 gene promoters for changes in methylation during postnatal liver development to test the hypothesis that developmental changes in methylation and expression are temporally correlated. We identified 31 genes that gained methylation and 111 that lost methylation from embryonic day 17.5 to postnatal day 21. Promoters undergoing methylation changes in postnatal liver tended not to be associated with CpG islands. At most genes studied, developmental changes in promoter methylation were associated with expression changes, suggesting both that transcriptional inactivity attracts de novo methylation, and that transcriptional activity can override DNA methylation and successively induce developmental hypomethylation. These in vivo data clearly indicate a role for DNA methylation in mammalian differentiation, and provide the novel insight that critical windows in mammalian developmental epigenetics extend well beyond early embryonic development.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Liver/growth & development , Mice/genetics , Animals , Female , Liver/embryology , Liver/metabolism , Male , Mice/embryology , Mice/growth & development , Mice/metabolism , Promoter Regions, Genetic
7.
Endocrinology ; 149(11): 5713-23, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617606

ABSTRACT

The periosteum is now widely recognized as a homeostatic and therapeutic target for actions of sex steroids and intermittent PTH administration. The mechanisms by which estrogens suppress but PTH promotes periosteal expansion are not known. In this report, we show that intermittent PTH(1-34) promotes differentiation of periosteal osteoblast precursors as evidenced by the stimulation of the expression or activity of alkaline phosphatase as well as of targets of the bone morphogenetic protein 2 (BMP-2) and Wnt pathways. In contrast, 17beta-estradiol (E2) had no effect by itself. However, it attenuated PTH- or BMP-2-induced differentiation of primary periosteal osteoblast progenitors. Administration of intermittent PTH to ovariectomized mice induced rapid phosphorylation of the BMP-2 target Smad1/5/8 in the periosteum. A replacement dose of E2 had no effect by itself but suppressed PTH-induced phosphorylation of Smad1/5/8. In contrast to its effects to stimulate periosteal osteoblast differentiation, PTH promoted and subsequently suppressed proliferation of periosteal osteoblast progenitors in vitro and in vivo. E2 promoted proliferation and attenuated the antiproliferative effect of PTH. Both hormones protected periosteal osteoblasts from apoptosis induced by various proapoptotic agents. These observations suggest that the different effects of PTH and estrogens on the periosteum result from opposing actions on the recruitment of early periosteal osteoblast progenitors. Intermittent PTH promotes osteoblast differentiation from periosteum-derived mesenchymal progenitors through ERK-, BMP-, and Wnt-dependent signaling pathways. Estrogens promote proliferation of early osteoblast progenitors but inhibit their differentiation by osteogenic agents such as PTH or BMP-2.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Estrogens/pharmacology , Osteoblasts/physiology , Parathyroid Hormone/pharmacology , Periosteum/cytology , Stem Cells/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Drug Administration Schedule , Estrogens/administration & dosage , Gonadal Steroid Hormones/pharmacology , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Parathyroid Hormone/administration & dosage , Periodicity , Periosteum/drug effects , Periosteum/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
8.
Genes Dev ; 20(21): 2937-42, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17050674

ABSTRACT

The perichondrium, a structure made of undifferentiated mesenchymal cells surrounding growth plate cartilage, regulates chondrocyte maturation through poorly understood mechanisms. Analyses of loss- and gain-of-function models show that Twist-1, whose expression in cartilage is restricted to perichondrium, favors chondrocyte maturation in a Runx2-dependent manner. Runx2, in turn, enhances perichondrial expression of Fgf18, a regulator of chondrocyte maturation. Accordingly, compound heterozygous embryos for Runx2 and Fgf18 deletion display the same chondrocyte maturation phenotype as Fgf18-null embryos. This study identifies a transcriptional basis for the inhibition of chondrocyte maturation by perichondrium and reveals that Runx2 fulfills antagonistic functions during chondrogenesis.


Subject(s)
Chondrogenesis/genetics , Core Binding Factor Alpha 1 Subunit/physiology , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/physiology , Core Binding Factor Alpha 1 Subunit/genetics , Fibroblast Growth Factors/metabolism , Gene Deletion , Growth Plate/embryology , Growth Plate/metabolism , Mesoderm/cytology , Mice , Mice, Mutant Strains , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...