Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Curr Osteoporos Rep ; 22(1): 80-95, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38198032

RESUMEN

PURPOSE OF THE REVIEW: The bone and hematopoietic tissues coemerge during development and are functionally intertwined throughout mammalian life. Oncostatin M (OSM) is an inflammatory cytokine of the interleukin-6 family produced by osteoblasts, bone marrow macrophages, and neutrophils. OSM acts via two heterodimeric receptors comprising GP130 with either an OSM receptor (OSMR) or a leukemia inhibitory factor receptor (LIFR). OSMR is expressed on osteoblasts, mesenchymal, and endothelial cells and mice deficient for the Osm or Osmr genes have both bone and blood phenotypes illustrating the importance of OSM and OSMR in regulating these two intertwined tissues. RECENT FINDINGS: OSM regulates bone mass through signaling via OSMR, adaptor protein SHC1, and transducer STAT3 to both stimulate osteoclast formation and promote osteoblast commitment; the effect on bone formation is also supported by action through LIFR. OSM produced by macrophages is an important inducer of neurogenic heterotopic ossifications in peri-articular muscles following spinal cord injury. OSM produced by neutrophils in the bone marrow induces hematopoietic stem and progenitor cell proliferation in an indirect manner via OSMR expressed by bone marrow stromal and endothelial cells that form hematopoietic stem cell niches. OSM acts as a brake to therapeutic hematopoietic stem cell mobilization in response to G-CSF and CXCR4 antagonist plerixafor. Excessive OSM production by macrophages in the bone marrow is a key contributor to poor hematopoietic stem cell mobilization (mobilopathy) in people with diabetes. OSM and OSMR may also play important roles in the progression of several cancers. It is increasingly clear that OSM plays unique roles in regulating the maintenance and regeneration of bone, hematopoietic stem and progenitor cells, inflammation, and skeletal muscles. Dysregulated OSM production can lead to bone pathologies, defective muscle repair and formation of heterotopic ossifications in injured muscles, suboptimal mobilization of hematopoietic stem cells, exacerbated inflammatory responses, and anti-tumoral immunity. Ongoing research will establish whether neutralizing antibodies or cytokine traps may be useful to correct pathologies associated with excessive OSM production.


Asunto(s)
Compuestos Heterocíclicos , Osificación Heterotópica , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Mamíferos/metabolismo , Oncostatina M/genética , Oncostatina M/metabolismo , Oncostatina M/farmacología
2.
Annu Rev Physiol ; 82: 507-529, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31553686

RESUMEN

Bone remodeling is essential for the repair and replacement of damaged and old bone. The major principle underlying this process is that osteoclast-mediated resorption of a quantum of bone is followed by osteoblast precursor recruitment; these cells differentiate to matrix-producing osteoblasts, which form new bone to replace what was resorbed. Evidence from osteopetrotic syndromes indicate that osteoclasts not only resorb bone, but also provide signals to promote bone formation. Osteoclasts act upon osteoblast lineage cells throughout their differentiation by facilitating growth factor release from resorbed matrix, producing secreted proteins and microvesicles, and expressing membrane-bound factors. These multiple mechanisms mediate the coupling of bone formation to resorption in remodeling. Additional interactions of osteoclasts with osteoblast lineage cells, including interactions with canopy and reversal cells, are required to achieve coordination between bone formation and resorption during bone remodeling.


Asunto(s)
Osteoblastos/fisiología , Osteoclastos/fisiología , Transducción de Señal/fisiología , Animales , Remodelación Ósea/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Exosomas/fisiología , Humanos , Osteogénesis
3.
J Cell Mol Med ; 26(14): 4021-4031, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35701367

RESUMEN

The inherited brittle bone disease osteogenesis imperfecta (OI) is commonly caused by COL1A1 and COL1A2 mutations that disrupt the collagen I triple helix. This causes intracellular endoplasmic reticulum (ER) retention of the misfolded collagen and can result in a pathological ER stress response. A therapeutic approach to reduce this toxic mutant load could be to stimulate mutant collagen degradation by manipulating autophagy and/or ER-associated degradation. Since carbamazepine (CBZ) both stimulates autophagy of misfolded collagen X and improves skeletal pathology in a metaphyseal chondrodysplasia model, we tested the effect of CBZ on bone structure and strength in 3-week-old male OI Col1a2 +/p.G610C and control mice. Treatment for 3 or 6 weeks with CBZ, at the dose effective in metaphyseal chondrodysplasia, provided no therapeutic benefit to Col1a2 +/p.G610C mouse bone structure, strength or composition, measured by micro-computed tomography, three point bending tests and Fourier-transform infrared microspectroscopy. In control mice, however, CBZ treatment for 6 weeks impaired femur growth and led to lower femoral cortical and trabecular bone mass. These data, showing the negative impact of CBZ treatment on the developing mouse bones, raise important issues which must be considered in any human clinical applications of CBZ in growing individuals.


Asunto(s)
Osteogénesis Imperfecta , Animales , Carbamazepina/farmacología , Carbamazepina/uso terapéutico , Colágeno/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Mutación/genética , Osteogénesis , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/metabolismo , Microtomografía por Rayos X
4.
Cell Mol Life Sci ; 78(15): 5755-5773, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34196732

RESUMEN

Cortical bone structure is a crucial determinant of bone strength, yet for many years studies of novel genes and cell signalling pathways regulating bone strength have focused on the control of trabecular bone mass. Here we focus on mechanisms responsible for cortical bone development, growth, and degeneration, and describe some recently described genetic-driven modifications in humans and mice that reveal how these processes may be controlled. We start with embryonic osteogenesis of preliminary bone structures preceding the cortex and describe how this structure consolidates then matures to a dense, vascularised cortex containing an increasing proportion of lamellar bone. These processes include modelling-induced, and load-dependent, asymmetric cortical expansion, which enables the cortex's transition from a highly porous woven structure to a consolidated and thickened highly mineralised lamellar bone structure, infiltrated by vascular channels. Sex-specific differences emerge during this process. With aging, the process of consolidation reverses: cortical pores enlarge, leading to greater cortical porosity, trabecularisation and loss of bone strength. Each process requires co-ordination between bone formation, bone mineralisation, vascularisation, and bone resorption, with a need for locational-, spatial- and cell-specific signalling pathways to mediate this co-ordination. We will discuss these processes, and a number of cell-signalling pathways identified in both murine and human genetic studies to regulate cortical bone mass, including signalling through gp130, STAT3, PTHR1, WNT16, NOTCH, NOTUM and sFRP4.


Asunto(s)
Desarrollo Óseo/genética , Condrocitos/fisiología , Hueso Cortical/crecimiento & desarrollo , Osteoblastos/fisiología , Osteoclastos/fisiología , Osteocitos/fisiología , Animales , Hueso Cortical/fisiología , Humanos , Porosidad , Transducción de Señal/genética
5.
Hum Mol Genet ; 28(15): 2573-2588, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31009951

RESUMEN

Mutations in subunits of the cilia-specific cytoplasmic dynein-2 (CD2) complex cause short-rib thoracic dystrophy syndromes (SRTDs), characterized by impaired bone growth and life-threatening perinatal respiratory complications. Different SRTD mutations result in varying disease severities. It remains unresolved whether this reflects the extent of retained hypomorphic protein functions or relative importance of the affected subunits for the activity of the CD2 holoenzyme. To define the contribution of the LC8-type dynein light chain subunit to the CD2 complex, we have generated Dynll1-deficient mouse strains, including the first-ever conditional knockout (KO) mutant for any CD2 subunit. Germline Dynll1 KO mice exhibit a severe ciliopathy-like phenotype similar to mice lacking another CD2 subunit, Dync2li1. Limb mesoderm-specific loss of Dynll1 results in severe bone shortening similar to human SRTD patients. Mechanistically, loss of Dynll1 leads to a partial depletion of other SRTD-related CD2 subunits, severely impaired retrograde intra-flagellar transport, significant thickening of primary cilia and cilia signaling defects. Interestingly, phenotypes of Dynll1-deficient mice are very similar to entirely cilia-deficient Kif3a/Ift88-null mice, except that they never present with polydactyly and retain relatively higher signaling outputs in parts of the hedgehog pathway. Compared to complete loss of Dynll1, maintaining very low DYNLL1 levels in mice lacking the Dynll1-transcription factor ASCIZ (ATMIN) results in significantly attenuated phenotypes and improved CD2 protein levels. The results suggest that primary cilia can maintain some functionality in the absence of intact CD2 complexes and provide a viable animal model for the analysis of the underlying bone development defects of SRTDs.


Asunto(s)
Enfermedades del Desarrollo Óseo/metabolismo , Cilios/metabolismo , Ciliopatías/metabolismo , Dineínas Citoplasmáticas/genética , Osteogénesis , Animales , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/fisiopatología , Células Cultivadas , Cilios/fisiología , Ciliopatías/genética , Ciliopatías/fisiopatología , Dineínas Citoplasmáticas/metabolismo , Dineínas Citoplasmáticas/fisiología , Extremidades/patología , Extremidades/fisiopatología , Proteínas Hedgehog/metabolismo , Masculino , Ratones , Ratones Noqueados , Fenotipo , Transducción de Señal , Factores de Transcripción/metabolismo
6.
Cytokine ; 146: 155655, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34332274

RESUMEN

The IL-6 family of cytokines comprises a large group of cytokines that all act via the formation of a signaling complex that includes the glycoprotein 130 (gp130) receptor. Despite this, many of these cytokines have unique roles that regulate the activity of bone forming osteoblasts, bone resorbing osteoclasts, bone-resident osteocytes, and cartilage cells (chondrocytes). These include specific functions in craniofacial development, longitudinal bone growth, and the maintenance of trabecular and cortical bone structure, and have been implicated in musculoskeletal pathologies such as craniosynostosis, osteoporosis, rheumatoid arthritis, osteoarthritis, and heterotopic ossifications. This review will work systematically through each member of this family and provide an overview and an update on the expression patterns and functions of each of these cytokines in the skeleton, as well as their negative feedback pathways, particularly suppressor of cytokine signaling 3 (SOCS3). The specific cytokines described are interleukin 6 (IL-6), interleukin 11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin 1 (CT-1), ciliary neurotrophic factor (CNTF), cardiotrophin-like cytokine factor 1 (CLCF1), neuropoietin, humanin and interleukin 27 (IL-27).


Asunto(s)
Huesos/anatomía & histología , Huesos/fisiología , Interleucina-6/metabolismo , Animales , Remodelación Ósea/fisiología , Retroalimentación Fisiológica , Humanos , Transducción de Señal
7.
Curr Osteoporos Rep ; 19(6): 604-615, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34757588

RESUMEN

PURPOSE OF THE REVIEW: Osteocytes are cells embedded within the bone matrix, but their function and specific patterns of gene expression remain only partially defined; this is beginning to change with recent studies using transcriptomics. This unbiased approach can generate large amounts of data and is now being used to identify novel genes and signalling pathways within osteocytes both at baseline conditions and in response to stimuli. This review outlines the methods used to isolate cell populations containing osteocytes, and key recent transcriptomic studies that used osteocyte-containing preparations from bone tissue. RECENT FINDINGS: Three common methods are used to prepare samples to examine osteocyte gene expression: digestion followed by sorting, laser capture microscopy, and the isolation of cortical bone shafts. All these methods present challenges in interpreting the data generated. Genes previously not known to be expressed by osteocytes have been identified and variations in osteocyte gene expression have been reported with age, sex, anatomical location, mechanical loading, and defects in bone strength. A substantial proportion of newly identified transcripts in osteocytes remain functionally undefined but several have been cross-referenced with functional data. Future work and improved methods (e.g. scRNAseq) likely provide useful resources for the study of osteocytes and important new information on the identity and functions of this unique cell type within the skeleton.


Asunto(s)
Huesos/citología , Huesos/metabolismo , Técnicas de Cultivo de Célula/métodos , Expresión Génica , Osteocitos/citología , Osteocitos/metabolismo , Humanos , Transcriptoma
8.
J Biol Chem ; 294(19): 7850-7863, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30923130

RESUMEN

Interleukin 6 (IL-6) supports development of bone-resorbing osteoclasts by acting early in the osteoblast lineage via membrane-bound (cis) or soluble (trans) receptors. Here, we investigated how IL-6 signals and modifies gene expression in differentiated osteoblasts and osteocytes and determined whether these activities can promote bone formation or support osteoclastogenesis. Moreover, we used a genetically altered mouse with circulating levels of the pharmacological IL-6 trans-signaling inhibitor sgp130-Fc to determine whether IL-6 trans-signaling is required for normal bone growth and remodeling. We found that IL-6 increases suppressor of cytokine signaling 3 (Socs3) and CCAAT enhancer-binding protein δ (Cebpd) mRNA levels and promotes signal transducer and activator of transcription 3 (STAT3) phosphorylation by both cis- and trans-signaling in cultured osteocytes. In contrast, RANKL (Tnfsf11) mRNA levels were elevated only by trans-signaling. Furthermore, we observed soluble IL-6 receptor release and ADAM metallopeptidase domain 17 (ADAM17) sheddase expression by osteocytes. Despite the observation that IL-6 cis-signaling occurs, IL-6 stimulated bone formation in vivo only via trans-signaling. Although IL-6 stimulated RANKL (Tnfsf11) mRNA in osteocytes, these cells did not support osteoclast formation in response to IL-6 alone; binucleated TRAP+ cells formed, and only in response to trans-signaling. Finally, pharmacological, sgp130-Fc-mediated inhibition of IL-6 trans-signaling did not impair bone growth or remodeling unless mice had circulating sgp130-Fc levels > 10 µg/ml. At those levels, osteopenia and impaired bone growth occurred, reducing bone strength. We conclude that high sgp130-Fc levels may have detrimental off-target effects on the skeleton.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Interleucina-6/metabolismo , Osteoclastos/metabolismo , Osteocitos/metabolismo , Osteogénesis , Transducción de Señal , Proteína ADAM17/metabolismo , Animales , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Ratones , Ligando RANK/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
9.
J Immunol ; 200(2): 632-642, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29212906

RESUMEN

A link between inflammatory disease and bone loss is now recognized. However, limited data exist on the impact of virus infection on bone loss and regeneration. Bone loss results from an imbalance in remodeling, the physiological process whereby the skeleton undergoes continual cycles of formation and resorption. The specific molecular and cellular mechanisms linking virus-induced inflammation to bone loss remain unclear. In the current study, we provide evidence that infection of mice with either lymphocytic choriomeningitis virus (LCMV) or pneumonia virus of mice (PVM) resulted in rapid and substantial loss of osteoblasts from the bone surface. Osteoblast ablation was associated with elevated levels of circulating inflammatory cytokines, including TNF-α, IFN-γ, IL-6, and CCL2. Both LCMV and PVM infections resulted in reduced osteoblast-specific gene expression in bone, loss of osteoblasts, and reduced serum markers of bone formation, including osteocalcin and procollagen type 1 N propeptide. Infection of Rag-1-deficient mice (which lack adaptive immune cells) or specific depletion of CD8+ T lymphocytes limited osteoblast loss associated with LCMV infection. By contrast, CD8+ T cell depletion had no apparent impact on osteoblast ablation in association with PVM infection. In summary, our data demonstrate dramatic loss of osteoblasts in response to virus infection and associated systemic inflammation. Further, the inflammatory mechanisms mediating viral infection-induced bone loss depend on the specific inflammatory condition.


Asunto(s)
Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Neumonía Murina/inmunología , Osteoblastos/virología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Animales , Biomarcadores , Médula Ósea/patología , Huesos/metabolismo , Huesos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Proteínas de Homeodominio/genética , Depleción Linfocítica , Ratones , Ratones Noqueados , Osteoblastos/inmunología , Osteogénesis
10.
Handb Exp Pharmacol ; 262: 1-25, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006260

RESUMEN

Bone tissue is comprised of a collagen-rich matrix containing non-collagenous organic compounds, strengthened by mineral crystals. Bone strength reflects the amount and structure of bone, as well as its quality. These qualities are determined and maintained by osteoblasts (bone-forming cells) and osteoclasts (bone-resorbing cells) on the surface of the bone and osteocytes embedded within the bone matrix. Bone development and growth also involves cartilage cells (chondrocytes). These cells do not act in isolation, but function in a coordinated manner, including co-ordination within each lineage, between the cells of bone, and between these cells and other cell types within the bone microenvironment. This chapter will briefly outline the cells of bone, their major functions, and some communication pathways responsible for controlling bone development and remodeling.


Asunto(s)
Osteoblastos , Osteoclastos , Remodelación Ósea , Huesos/fisiopatología , Osteocitos/fisiología
11.
J Cell Mol Med ; 23(3): 1735-1745, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30597759

RESUMEN

Osteogenesis imperfecta (OI) is commonly caused by heterozygous type I collagen structural mutations that disturb triple helix folding and integrity. This mutant-containing misfolded collagen accumulates in the endoplasmic reticulum (ER) and induces a form of ER stress associated with negative effects on osteoblast differentiation and maturation. Therapeutic induction of autophagy to degrade the mutant collagens could therefore be useful in ameliorating the ER stress and deleterious downstream consequences. To test this, we treated a mouse model of mild to moderate OI (α2(I) G610C) with dietary rapamycin from 3 to 8 weeks of age and effects on bone mass and mechanical properties were determined. OI bone mass and mechanics were, as previously reported, compromised compared to WT. While rapamycin treatment improved the trabecular parameters of WT and OI bones, the biomechanical deficits of OI bones were not rescued. Importantly, we show that rapamycin treatment suppressed the longitudinal and transverse growth of OI, but not WT, long bones. Our work demonstrates that dietary rapamycin offers no clinical benefit in this OI model and furthermore, the impact of rapamycin on OI bone growth could exacerbate the clinical consequences during periods of active bone growth in patients with OI caused by collagen misfolding mutations.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Colágeno Tipo I/fisiología , Modelos Animales de Enfermedad , Inmunosupresores/farmacología , Osteoblastos/efectos de los fármacos , Osteogénesis Imperfecta/tratamiento farmacológico , Sirolimus/farmacología , Animales , Apoptosis , Cadena alfa 1 del Colágeno Tipo I , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoblastos/citología , Osteogénesis , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología
12.
Development ; 143(4): 648-57, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26755702

RESUMEN

The majority of the skeleton arises by endochondral ossification, whereby cartilaginous templates expand and are resorbed by osteoclasts then replaced by osteoblastic bone formation. Ephrin B2 is a receptor tyrosine kinase expressed by osteoblasts and growth plate chondrocytes that promotes osteoblast differentiation and inhibits osteoclast formation. We investigated the role of ephrin B2 in endochondral ossification using Osx1Cre-targeted gene deletion. Neonatal Osx1Cre.Efnb2(Δ/Δ) mice exhibited a transient osteopetrosis demonstrated by increased trabecular bone volume with a high content of growth plate cartilage remnants and increased cortical thickness, but normal osteoclast numbers within the primary spongiosa. Osteoclasts at the growth plate had an abnormal morphology and expressed low levels of tartrate-resistant acid phosphatase; this was not observed in more mature bone. Electron microscopy revealed a lack of sealing zones and poor attachment of Osx1Cre.Efnb2(Δ/Δ) osteoclasts to growth plate cartilage. Osteoblasts at the growth plate were also poorly attached and impaired in their ability to deposit osteoid. By 6 months of age, trabecular bone mass, osteoclast morphology and osteoid deposition by Osx1Cre.Efnb2(Δ/Δ) osteoblasts were normal. Cultured chondrocytes from Osx1Cre.Efnb2(Δ/Δ) neonates showed impaired support of osteoclastogenesis but no significant change in Rankl (Tnfsf11) levels, whereas Adamts4 levels were significantly reduced. A population of ADAMTS4(+) early hypertrophic chondrocytes seen in controls was absent from Osx1Cre.Efnb2(Δ/Δ) neonates. This suggests that Osx1Cre-expressing cells, including hypertrophic chondrocytes, are dependent on ephrin B2 for their production of cartilage-degrading enzymes, including ADAMTS4, and this might be required for attachment of osteoclasts and osteoblasts to the cartilage surface during endochondral ossification.


Asunto(s)
Cartílago/patología , Condrocitos/metabolismo , Efrina-B2/metabolismo , Osteoclastos/patología , Osteogénesis , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Animales , Animales Recién Nacidos , Cartílago/metabolismo , Adhesión Celular , Diferenciación Celular , Condrocitos/patología , Femenino , Regulación de la Expresión Génica , Inmunohistoquímica , Integrasas/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Tamaño de los Órganos , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/ultraestructura , Osteogénesis/genética , Osteopetrosis/genética , Osteopetrosis/patología , Fenotipo , Procolágeno N-Endopeptidasa/metabolismo , Tibia/metabolismo , Tibia/patología
13.
Curr Osteoporos Rep ; 17(5): 270-280, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31401710

RESUMEN

PURPOSE OF REVIEW: We outline the diverse processes contributing to bone mineralization and bone matrix maturation by describing two mouse models with bone strength defects caused by restricted deletion of the receptor tyrosine kinase ligand EphrinB2. RECENT FINDINGS: Stage-specific EphrinB2 deletion differs in its effects on skeletal strength. Early-stage deletion in osteoblasts leads to osteoblast apoptosis, delayed initiation of mineralization, and increased bone flexibility. Deletion later in the lineage targeted to osteocytes leads to a brittle bone phenotype and increased osteocyte autophagy. In these latter mice, although mineralization is initiated normally, all processes involved in matrix maturation, including mineral accrual, carbonate substitution, and collagen compaction, progress more rapidly. Osteoblasts and osteocytes control the many processes involved in bone mineralization; defining the contributing signaling activities may lead to new ways to understand and treat human skeletal fragilities.


Asunto(s)
Calcificación Fisiológica/fisiología , Efrina-B2/antagonistas & inhibidores , Osteoblastos/fisiología , Osteocitos/fisiología , Animales , Biomarcadores/metabolismo , Efrina-B2/metabolismo , Ratones , Modelos Animales , Fenotipo , Transducción de Señal
14.
Calcif Tissue Int ; 103(6): 625-637, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30019315

RESUMEN

Bone is formed by deposition of a collagen-containing matrix (osteoid) that hardens over time as mineral crystals accrue and are modified; this continues until bone remodeling renews that site. Pharmacological agents for osteoporosis differ in their effects on bone remodeling, and we hypothesized that they may differently modify bone mineral accrual. We, therefore, assessed newly formed bone in mature ovariectomized rabbits treated with the anti-resorptive bisphosphonate alendronate (ALN-100µ g/kg/2×/week), the anabolic parathyroid hormone (PTH (1-34)-15µ g/kg/5×/week), or the experimental anti-resorptive odanacatib (ODN 7.5 µM/day), which suppresses bone resorption without suppressing bone formation. Treatments were administered for 10 months commencing 6 months after ovariectomy (OVX). Strength testing, histomorphometry, and synchrotron Fourier-transform infrared microspectroscopy were used to measure bone strength, bone formation, and mineral accrual, respectively, in newly formed endocortical and intracortical bone. In Sham and OVX endocortical and intracortical bone, three modifications occurred as the bone matrix aged: mineral accrual (increase in mineral:matrix ratio), carbonate substitution (increase in carbonate:mineral ratio), and collagen molecular compaction (decrease in amide I:II ratio). ALN suppressed bone formation but mineral accrued normally at those sites where bone formation occurred. PTH stimulated bone formation on endocortical, periosteal, and intracortical bone surfaces, but mineral accrual and carbonate substitution were suppressed, particularly in intracortical bone. ODN treatment did not suppress bone formation, but newly deposited endocortical bone matured more slowly with ODN, and ODN-treated intracortical bone had less carbonate substitution than controls. In conclusion, these agents differ in their effects on the bone matrix. While ALN suppresses bone formation, it does not modify bone mineral accrual in endocortical or intracortical bone. While ODN does not suppress bone formation, it slows matrix maturation. PTH stimulates modelling-based bone formation not only on endocortical and trabecular surfaces, but may also do so in intracortical bone; at this site, new bone deposited contains less mineral than normal.


Asunto(s)
Alendronato/farmacología , Compuestos de Bifenilo/farmacología , Conservadores de la Densidad Ósea/farmacología , Huesos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Animales , Remodelación Ósea/efectos de los fármacos , Huesos/fisiología , Calcificación Fisiológica/efectos de los fármacos , Femenino , Osteogénesis/efectos de los fármacos , Ovariectomía , Conejos
15.
Neuroendocrinology ; 107(4): 340-354, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30208370

RESUMEN

BACKGROUND/AIMS: Abuse of toluene products (e.g., glue-sniffing) primarily occurs during adolescence and has been associated with appetite suppression and weight impairments. However, the metabolic phenotype arising from adolescent inhalant abuse has never been fully characterised, and its persistence during abstinence and underlying mechanisms remain unknown. METHODS: Adolescent male Wistar rats (post-natal day 27) were exposed to inhaled toluene (10,000 ppm) (n = 32) or air (n = 48) for 1 h/day, 3 days/week for 4 weeks, followed by 4 weeks of abstinence. Twenty air rats were pair-fed to the toluene group, to differentiate the direct effects of toluene from under-nutrition. Food intake, weight, and growth were monitored. Metabolic hormones were measured after exposure and abstinence periods. Energy expenditure was measured using indirect calorimetry. Adrenal function was assessed using adrenal histology and hormone testing. RESULTS: Inhalant abuse suppressed appetite and increased energy expenditure. Reduced weight gain and growth were observed in both the toluene and pair-fed groups. Compared to the pair-fed group, and despite normalisation of food intake, the suppression of weight and growth for toluene-exposed rats persisted during abstinence. After exposure, toluene-exposed rats had low fasting blood glucose and insulin compared to the air and pair-fed groups. Consistent with adrenal insufficiency, adrenal hypertrophy and increased basal adrenocorticotropic hormone were observed in the toluene-exposed rats, despite normal basal corticosterone levels. CONCLUSIONS: Inhalant abuse results in negative energy balance, persistent growth impairment, and endocrine changes suggestive of adrenal insufficiency. We conclude that adrenal insufficiency contributes to the negative energy balance phenotype, potentially presenting a significant additional health risk for inhalant users.


Asunto(s)
Enfermedades de las Glándulas Suprarrenales/inducido químicamente , Trastornos del Crecimiento/inducido químicamente , Abuso de Inhalantes/complicaciones , Enfermedades Metabólicas/inducido químicamente , Maduración Sexual , Adolescente , Conducta del Adolescente/efectos de los fármacos , Conducta del Adolescente/fisiología , Desarrollo del Adolescente/efectos de los fármacos , Enfermedades de las Glándulas Suprarrenales/metabolismo , Enfermedades de las Glándulas Suprarrenales/fisiopatología , Glándulas Suprarrenales/fisiopatología , Animales , Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/fisiopatología , Humanos , Abuso de Inhalantes/metabolismo , Abuso de Inhalantes/patología , Abuso de Inhalantes/fisiopatología , Masculino , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/fisiopatología , Actividad Motora/efectos de los fármacos , Fenotipo , Ratas , Ratas Wistar , Maduración Sexual/efectos de los fármacos , Maduración Sexual/fisiología , Tolueno/toxicidad
16.
PLoS Genet ; 11(4): e1005160, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25859855

RESUMEN

RECQL4 mutations are associated with Rothmund Thomson Syndrome (RTS), RAPADILINO Syndrome and Baller-Gerold Syndrome. These patients display a range of benign skeletal abnormalities such as low bone mass. In addition, RTS patients have a highly increased incidence of osteosarcoma (OS). The role of RECQL4 in normal adult bone development and homeostasis is largely uncharacterized and how mutation of RECQL4 contributes to OS susceptibility is not known. We hypothesised that Recql4 was required for normal skeletal development and both benign and malignant osteoblast function, which we have tested in the mouse. Recql4 deletion in vivo at the osteoblastic progenitor stage of differentiation resulted in mice with shorter bones and reduced bone volume, assessed at 9 weeks of age. This was associated with an osteoblast intrinsic decrease in mineral apposition rate and bone formation rate in the Recql4-deficient cohorts. Deletion of Recql4 in mature osteoblasts/osteocytes in vivo, however, did not cause a detectable phenotype. Acute deletion of Recql4 in primary osteoblasts or shRNA knockdown in an osteoblastic cell line caused failed proliferation, accompanied by cell cycle arrest, induction of apoptosis and impaired differentiation. When cohorts of animals were aged long term, the loss of Recql4 alone was not sufficient to initiate OS. We then crossed the Recql4fl/fl allele to a fully penetrant OS model (Osx-Cre p53fl/fl). Unexpectedly, the Osx-Cre p53fl/flRecql4fl/fl (dKO) animals had a significantly increased OS-free survival compared to Osx-Cre p53fl/fl or Osx-Cre p53fl/flRecql4fl/+ (het) animals. The extended survival was explained when the Recql4 status in the tumors that arose was assessed, and in no case was there complete deletion of Recql4 in the dKO OS. These data provide a mechanism for the benign skeletal phenotypes of RECQL4 mutation syndromes. We propose that tumor suppression and osteosarcoma susceptibility are most likely a function of mutant, not null, alleles of RECQL4.


Asunto(s)
Neoplasias Óseas/genética , Osteoblastos/metabolismo , Osteogénesis , Osteosarcoma/genética , RecQ Helicasas/metabolismo , Animales , Neoplasias Óseas/metabolismo , Proliferación Celular , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/metabolismo , RecQ Helicasas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
J Biol Chem ; 291(41): 21703-21716, 2016 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-27539849

RESUMEN

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are IL-6 family members with a wide range of biological functions. Human OSM (hOSM) and murine LIF (mLIF) act in mouse cells via a LIF receptor (LIFR)-glycoprotein 130 (gp130) heterodimer. In contrast, murine OSM (mOSM) signals mainly via an OSM receptor (OSMR)-gp130 heterodimer and binds with only very low affinity to mLIFR. hOSM and mLIF stimulate bone remodeling by both reducing osteocytic sclerostin and up-regulating the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) in osteoblasts. In the absence of OSMR, mOSM still strongly suppressed sclerostin and stimulated bone formation but did not induce RANKL, suggesting that intracellular signaling activated by the low affinity interaction of mOSM with mLIFR is different from the downstream effects when mLIF or hOSM interacts with the same receptor. Both STAT1 and STAT3 were activated by mOSM in wild type cells or by mLIF/hOSM in wild type and Osmr-/- cells. In contrast, in Osmr-/- primary osteocyte-like cells stimulated with mOSM (therefore acting through mLIFR), microarray expression profiling and Western blotting analysis identified preferential phosphorylation of STAT3 and induction of its target genes but not of STAT1 and its target genes; this correlated with reduced phosphorylation of both gp130 and LIFR. In a mouse model of spontaneous osteopenia caused by hyperactivation of STAT1/3 signaling downstream of gp130 (gp130Y757F/Y757F), STAT1 deletion rescued the osteopenic phenotype, indicating a beneficial effect of promoting STAT3 signaling over STAT1 downstream of gp130 in this low bone mass condition, and this may have therapeutic value.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Oncostatina M/metabolismo , Osteocitos/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Huesos/metabolismo , Huesos/patología , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Ratones , Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Tamaño de los Órganos , Osteocitos/patología , Fosforilación/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética
18.
J Biol Chem ; 291(4): 1631-1642, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26598522

RESUMEN

Parathyroid hormone (PTH) is an important regulator of osteoblast function and is the only anabolic therapy currently approved for treatment of osteoporosis. The PTH receptor (PTH1R) is a G protein-coupled receptor that signals via multiple G proteins including Gsα. Mice expressing a constitutively active mutant PTH1R exhibited a dramatic increase in trabecular bone that was dependent upon expression of Gsα in the osteoblast lineage. Postnatal removal of Gsα in the osteoblast lineage (P-Gsα(OsxKO) mice) yielded markedly reduced trabecular and cortical bone mass. Treatment with anabolic PTH(1-34) (80 µg/kg/day) for 4 weeks failed to increase trabecular bone volume or cortical thickness in male and female P-Gsα(OsxKO) mice. Surprisingly, in both male and female mice, PTH administration significantly increased osteoblast numbers and bone formation rate in both control and P-Gsα(OsxKO) mice. In mice that express a mutated PTH1R that activates adenylyl cyclase and protein kinase A (PKA) via Gsα but not phospholipase C via Gq/11 (D/D mice), PTH significantly enhanced bone formation, indicating that phospholipase C activation is not required for increased bone turnover in response to PTH. Therefore, although the anabolic effect of intermittent PTH treatment on trabecular bone volume is blunted by deletion of Gsα in osteoblasts, PTH can stimulate osteoblast differentiation and bone formation. Together these findings suggest that alternative signaling pathways beyond Gsα and Gq/11 act downstream of PTH on osteoblast differentiation.


Asunto(s)
Anabolizantes/administración & dosificación , Desarrollo Óseo/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Terapia de Reemplazo de Hormonas , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Hormona Paratiroidea/administración & dosificación , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Diferenciación Celular/efectos de los fármacos , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Osteoblastos/metabolismo , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
19.
Proc Natl Acad Sci U S A ; 111(16): 6040-5, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24733914

RESUMEN

Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.


Asunto(s)
Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Artritis/virología , Resorción Ósea/patología , Resorción Ósea/virología , Osteoblastos/patología , Virus del Río Ross/fisiología , Fosfatasa Ácida/sangre , Adulto , Infecciones por Alphavirus/sangre , Animales , Anticuerpos Neutralizantes/farmacología , Artritis/sangre , Artritis/patología , Resorción Ósea/sangre , Huesos/diagnóstico por imagen , Huesos/patología , Huesos/virología , Femenino , Placa de Crecimiento/efectos de los fármacos , Placa de Crecimiento/patología , Placa de Crecimiento/virología , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/biosíntesis , Isoenzimas/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Neutralización , Osteoblastos/efectos de los fármacos , Osteoblastos/virología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoclastos/virología , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Fenotipo , Ligando RANK/metabolismo , Virus del Río Ross/efectos de los fármacos , Líquido Sinovial/metabolismo , Fosfatasa Ácida Tartratorresistente , Replicación Viral/efectos de los fármacos , Microtomografía por Rayos X
20.
J Virol ; 89(1): 581-93, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339772

RESUMEN

UNLABELLED: The recent global resurgence of arthritogenic alphaviruses, in particular chikungunya virus (CHIKV), highlights an urgent need for the development of therapeutic intervention strategies. While there has been significant progress in defining the pathophysiology of alphaviral disease, relatively little is known about the mechanisms involved in CHIKV-induced arthritis or potential therapeutic options to treat the severe arthritic symptoms associated with infection. Here, we used microcomputed tomographic (µCT) and histomorphometric analyses to provide previously undescribed evidence of reduced bone volume in the proximal tibial epiphysis of CHIKV-infected mice compared to the results for mock controls. This was associated with a significant increase in the receptor activator of nuclear factor-κB ligand/osteoprotegerin (RANKL/OPG) ratio in infected murine joints and in the serum of CHIKV patients. The expression levels of the monocyte chemoattractant proteins (MCPs), including MCP-1/CCL2, MCP-2/CCL8, and MCP-3/CCL7, were also highly elevated in joints of CHIKV-infected mice, accompanied by increased cellularity within the bone marrow in tibial epiphysis and ankle joints. Both this effect and CHIKV-induced bone loss were significantly reduced by treatment with the MCP inhibitor bindarit. Collectively, these findings demonstrate a unique role for MCPs in promoting CHIKV-induced osteoclastogenesis and bone loss during disease and suggest that inhibition of MCPs with bindarit may be an effective therapy for patients affected with alphavirus-induced bone loss. IMPORTANCE: Arthritogenic alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), cause worldwide outbreaks of polyarthritis, which can persist in patients for months following infection. Previous studies have shown that host proinflammatory soluble factors are associated with CHIKV disease severity. Furthermore, it is established that chemokine (C-C motif) ligand 2 (CCL2/MCP-1) is important in cellular recruitment and inducing bone-resorbing osteoclast (OC) formation. Here, we show that CHIKV replicates in bone and triggers bone loss by increasing the RANKL/OPG ratio. CHIKV infection results in MCP-induced cellular infiltration in the inflamed joints, and bone loss can be ameliorated by treatment with an MCP-inhibiting drug, bindarit. Taken together, our data reveal a previously undescribed role for MCPs in CHIKV-induced bone loss: one of recruiting monocytes/OC precursors to joint sites and thereby favoring a pro-osteoclastic microenvironment. This suggests that bindarit may be an effective treatment for alphavirus-induced bone loss and arthritis in humans.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Resorción Ósea/prevención & control , Quimiocina CCL2/antagonistas & inhibidores , Fiebre Chikungunya/complicaciones , Indazoles/administración & dosificación , Propionatos/administración & dosificación , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA