Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Biol ; 26(14): 1843-53, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27426518

ABSTRACT

Circadian rhythms are deeply rooted in the biology of virtually all organisms. The pervasive use of artificial lighting in modern society disrupts circadian rhythms and can be detrimental to our health. To investigate the relationship between disrupting circadian rhythmicity and disease, we exposed mice to continuous light (LL) for 24 weeks and measured several major health parameters. Long-term neuronal recordings revealed that 24 weeks of LL reduced rhythmicity in the central circadian pacemaker of the suprachiasmatic nucleus (SCN) by 70%. Strikingly, LL exposure also reduced skeletal muscle function (forelimb grip strength, wire hanging duration, and grid hanging duration), caused trabecular bone deterioration, and induced a transient pro-inflammatory state. After the mice were returned to a standard light-dark cycle, the SCN neurons rapidly recovered their normal high-amplitude rhythm, and the aforementioned health parameters returned to normal. These findings strongly suggest that a disrupted circadian rhythm reversibly induces detrimental effects on multiple biological processes.


Subject(s)
Biological Clocks , Circadian Rhythm , Light , Mice/physiology , Photoperiod , Suprachiasmatic Nucleus/physiology , Animals , Male , Mice, Inbred C57BL
2.
Histochem Cell Biol ; 144(1): 1-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25850409

ABSTRACT

Osteocytes are the predominant cells in bone, where they form a cellular network and display important functions in bone homeostasis, phosphate metabolism and mechanical transduction. Several proteins strongly expressed by osteocytes are involved in these processes, e.g., sclerostin, DMP-1, PHEX, FGF23 and MEPE, while others are upregulated during differentiation of osteoblasts into osteocytes, e.g., osteocalcin and E11. The receptor-type protein tyrosine phosphatase µ (RPTPµ) has been described to be expressed in cells which display a cellular network, e.g., endothelial and neuronal cells, and is implied in mechanotransduction. In a capillary outgrowth assay using metatarsals derived from RPTPµ-knock-out/LacZ knock-in mice, we observed that the capillary structures grown out of the metatarsals were stained blue, as expected. Surprisingly, cells within the metatarsal bone tissue were positive for LacZ activity as well, indicating that RPTPµ is also expressed by osteocytes. Subsequent histochemical analysis showed that within bone, RPTPµ is expressed exclusively in early-stage osteocytes. Analysis of bone marrow cell cultures revealed that osteocytes are present in the nodules and an enzymatic assay enabled the quantification of the amount of osteocytes. No apparent bone phenotype was observed when tibiae of RPTPµ-knock-out/LacZ knock-in mice were analyzed by µCT at several time points during aging, although a significant reduction in cortical bone was observed in RPTPµ-knock-out/LacZ knock-in mice at 20 weeks. Changes in trabecular bone were more subtle. Our data show that RPTPµ is a new marker for osteocytes.


Subject(s)
Metatarsal Bones/cytology , Osteocytes/enzymology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Biomarkers , Bone Marrow Cells/enzymology , Bone and Bones/diagnostic imaging , Fibroblast Growth Factor-23 , Gene Knock-In Techniques , Histocytochemistry , Mechanotransduction, Cellular , Metatarsal Bones/growth & development , Mice , Mice, Knockout , Osteogenesis , Tomography, X-Ray Computed
3.
Endocrinology ; 155(11): 4287-95, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25060361

ABSTRACT

Ghrelin receptor-deficient (Ghsr-/-) mice that lack acylated ghrelin (AG) signaling retain a metabolic response to unacylated ghrelin (UAG). Recently, we showed that Ghsr-deficiency affects bone metabolism. The aim of this study was to further establish the impact of AG and UAG on bone metabolism. We compared bone metabolism in Ghsr-/- (lacking only AG signaling) and ghrelin-deficient (Ghrl-/-; both AG and UAG deficient) male mice. Ghrl-/- mice had lower cortical bone mass, whereas Ghsr-/- mice had lower trabecular bone mass. This demonstrates bone compartment-specific effects of AG and a role for UAG in bone metabolism. Also, Ghrl-/- but not Ghsr-/- mice had increased bone formation rate and increased osteogenic stem cell numbers in their bone marrow. In ex vivo bone marrow cultures both AG and UAG inhibited osteoblast differentiation. This indicated that bone resorption must be increased in these mice. Accordingly, osteoclastogenesis rate was faster in bone marrow cultures from Ghsr-/- and Ghrl-/- mice, and osteoclast formation was inhibited by AG signaling and partially suppressed by UAG. In osteoblast cultures, AG markedly induced osteoprotegerin gene expression and both peptides reduced RANKL/osteoprotegerin ratio. These data describe unique cell-type specific effects of AG and UAG within a single tissue, supporting a tight and complex control of bone formation and resorption as well as a link between nutrition and bone metabolism. The balance between AG and UAG actions in the bone marrow may lead to bone compartmental-specific effects.


Subject(s)
Bone Remodeling/genetics , Bone and Bones/metabolism , Ghrelin/genetics , Receptors, Ghrelin/genetics , Acylation , Animals , Bone Density/drug effects , Bone Density/genetics , Bone Remodeling/drug effects , Bone and Bones/anatomy & histology , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Cells, Cultured , Ghrelin/metabolism , Ghrelin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , X-Ray Microtomography
4.
Endocrinology ; 153(8): 3593-602, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700774

ABSTRACT

The mutual interplay between energy homeostasis and bone metabolism is an important emerging concept. Ghrelin and leptin antagonize each other in regulating energy balance, but the role of this interaction in bone metabolism is unknown. Using ghrelin receptor and leptin-deficient mice, we show that ghrelin has dual effects on osteoclastogenesis, inhibiting osteoclast progenitors directly and stimulating osteoclastogenesis via a more potent systemic/central pathway. Using mice with combined ghrelin receptor and leptin deficiency, we find that this systemic osteoclastogenic activity is suppressed by leptin, thus balancing the two counterregulatory ghrelin pathways and leading to an unchanged bone structure. With aging, this osteoclastogenic ghrelin pathway is lost, unmasking the direct protective effect of ghrelin on bone structure. In conclusion, we identify a novel regulatory network linking orexigenic and anorectic metabolic factors with bone metabolism that is age dependent.


Subject(s)
Bone Resorption/metabolism , Bone and Bones/metabolism , Ghrelin/metabolism , Leptin/metabolism , Age Factors , Animals , Bone Resorption/genetics , Ghrelin/genetics , Ghrelin/pharmacology , Leptin/genetics , Leptin/pharmacology , Male , Mice , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/drug effects , Real-Time Polymerase Chain Reaction
5.
Vitam Horm ; 77: 239-58, 2008.
Article in English | MEDLINE | ID: mdl-17983859

ABSTRACT

A consequence of gastrectomy is loss of bone mass. Several mechanisms have been proposed, such as malabsorption of vitamins and minerals. Additionally, a peptide hormone produced in the stomach has been shown to mediate a calcitropic effect on bone. The identity of this peptide has not been elucidated, but ghrelin, produced by A-like cells in the fundus of the stomach, could be a good candidate. Ghrelin stimulates growth hormone (GH) secretion both in vivo and in vitro, and could by this means have a positive effect on bone. There is also evidence for direct effects of ghrelin on bone. We discuss here the role that ghrelin may play in bone metabolism, based on the most recent literature.


Subject(s)
Bone and Bones/metabolism , Ghrelin/metabolism , Animals , Bone Remodeling/physiology , Gastrectomy/adverse effects , Humans , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...