Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Biol Chem ; 300(4): 107158, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38479598

RESUMEN

Single-cell RNA-seq has led to novel designations for mesenchymal cells associated with bone as well as multiple designations for what appear to be the same cell type. The main goals of this study were to increase the amount of single-cell RNA sequence data for osteoblasts and osteocytes, to compare cells from the periosteum to those inside bone, and to clarify the major categories of cell types associated with murine bone. We created an atlas of murine bone-associated cells by harmonizing published datasets with in-house data from cells targeted by Osx1-Cre and Dmp1-Cre driver strains. Cells from periosteal bone were analyzed separately from those isolated from the endosteum and trabecular bone. Over 100,000 mesenchymal cells were mapped to reveal 11 major clusters designated fibro-1, fibro-2, chondrocytes, articular chondrocytes, tenocytes, adipo-Cxcl12 abundant reticular (CAR), osteo-CAR, preosteoblasts, osteoblasts, osteocytes, and osteo-X, the latter defined in part by periostin expression. Osteo-X, osteo-CAR, and preosteoblasts were closely associated with osteoblasts at the trabecular bone surface. Wnt16 was expressed in multiple cell types from the periosteum but not in cells from endocortical or cancellous bone. Fibro-2 cells, which express markers of stem cells, localized to the periosteum but not trabecular bone in adult mice. Suppressing bone remodeling eliminated osteoblasts and altered gene expression in preosteoblasts but did not change the abundance or location of osteo-X or osteo-CAR cells. These results provide a framework for identifying bone cell types in murine single-cell RNA-seq datasets and suggest that osteoblast progenitors reside near the surface of remodeling bone.


Asunto(s)
Células Madre Mesenquimatosas , Osteoblastos , Osteocitos , Periostio , Animales , Ratones , Condrocitos/metabolismo , Condrocitos/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Osteoblastos/metabolismo , Osteoblastos/citología , Osteocitos/metabolismo , Osteocitos/citología , Periostio/citología , Periostio/metabolismo , Análisis de la Célula Individual , Ratones Endogámicos C57BL
2.
Physiol Rev ; 97(1): 135-187, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27807202

RESUMEN

Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.


Asunto(s)
Andrógenos/metabolismo , Huesos/metabolismo , Huesos/fisiopatología , Estrógenos/metabolismo , Osteoporosis/fisiopatología , Animales , Femenino , Homeostasis/fisiología , Humanos , Masculino , Osteoporosis/metabolismo
3.
Am J Pathol ; 190(12): 2436-2452, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32926855

RESUMEN

We identified a family with a UMOD gene mutation (C106F) resulting in glomerular inflammation and complement deposition. To determine if the observed phenotype is due to immune system activation by mutant uromodulin, a mouse strain with a homologous cysteine to phenylalanine mutation (C105F) in the UMOD gene was generated using CRISPR-Cas9 gene editing and the effect of this mutation on mononuclear phagocytic cells was examined. Mutant mice developed high levels of intracellular and secreted aggregated uromodulin, resulting in anti-uromodulin antibodies and circulating uromodulin containing immune complexes with glomerular deposition and kidney fibrosis with aging. F4/80+ and CD11c+ kidney cells phagocytize uromodulin. Differential gene expression analysis by RNA sequencing of F4/80+ phagocytic cells revealed activation of the activating transcription factor 5 (ATF5)-mediated stress response pathway in mutant mice. Phagocytosis of mutant uromodulin by cultured dendritic cells resulted in activation of the endoplasmic reticulum stress response pathway and markers of cell inactivation, an effect not seen with wild-type protein. Mutant mice demonstrate a twofold increase in T-regulatory cells, consistent with induction of immune tolerance, resulting in decreased inflammatory response and improved tissue repair following ischemia-reperfusion injury. The C105F mutation results in autoantibodies against aggregated misfolded protein with immune complex formation and kidney fibrosis. Aggregated uromodulin may induce dendritic cell tolerance following phagocytosis through an unfolded protein/endoplasmic reticulum stress response pathway, resulting in decreased inflammation following tissue injury.


Asunto(s)
Autoinmunidad/inmunología , Estrés del Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Fagocitos/inmunología , Uromodulina/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Fibrosis/patología , Riñón/inmunología , Riñón/patología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Ratones , Fenotipo , Respuesta de Proteína Desplegada/inmunología , Uromodulina/genética , Uromodulina/inmunología
4.
Hum Mol Genet ; 26(4): 686-701, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28040732

RESUMEN

The recent identification of profilin1 mutations in 25 familial ALS cases has linked altered function of this cytoskeleton-regulating protein to the pathogenesis of motor neuron disease. To investigate the pathological role of mutant profilin1 in motor neuron disease, we generated transgenic lines of mice expressing human profilin1 with a mutation at position 118 (hPFN1G118V). One of the mouse lines expressing high levels of mutant human PFN1 protein in the brain and spinal cord exhibited many key clinical and pathological features consistent with human ALS disease. These include loss of lower (ventral horn) and upper motor neurons (corticospinal motor neurons in layer V), mutant profilin1 aggregation, abnormally ubiquitinated proteins, reduced choline acetyltransferase (ChAT) enzyme expression, fragmented mitochondria, glial cell activation, muscle atrophy, weight loss, and reduced survival. Our investigations of actin dynamics and axonal integrity suggest that mutant PFN1 protein is associated with an abnormally low filamentous/globular (F/G)-actin ratio that may be the underlying cause of severe damage to ventral root axons resulting in a Wallerian-like degeneration. These observations indicate that our novel profilin1 mutant mouse line may provide a new ALS model with the opportunity to gain unique perspectives into mechanisms of neurodegeneration that contribute to ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Mutación Missense , Profilinas/biosíntesis , Médula Espinal/metabolismo , Sustitución de Aminoácidos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Profilinas/genética , Médula Espinal/patología
5.
Curr Osteoporos Rep ; 16(4): 458-465, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29802575

RESUMEN

PURPOSE OF REVIEW: The goal of this review is to highlight some of the considerations involved in creating animal models to study rare bone diseases and then to compare and contrast approaches to creating such models, focusing on the advantages and novel opportunities offered by the CRISPR-Cas system. RECENT FINDINGS: Gene editing after creation of double-stranded breaks in chromosomal DNA is increasingly being used to modify animal genomes. Multiple tools can be used to create such breaks, with the newest ones being based on the bacterial adaptive immune system known as CRISPR/Cas. Advances in gene editing have increased the ease and speed, while reducing the cost, of creating novel animal models of disease. Gene editing has also expanded the number of animal species in which genetic modification can be performed. These changes have significantly increased the options for investigators seeking to model rare bone diseases in animals.


Asunto(s)
Enfermedades Óseas/genética , Modelos Animales de Enfermedad , Edición Génica/métodos , Marcación de Gen/métodos , Enfermedades Raras/genética , Animales , Sistemas CRISPR-Cas , Calcio/deficiencia , Calcio de la Dieta , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Roturas del ADN de Doble Cadena , Proteínas de la Matriz Extracelular , Interacción Gen-Ambiente , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Chaperonas Moleculares , Osteítis Deformante/genética , Osteítis Deformante/metabolismo , Osteogénesis Imperfecta/genética , Proteínas/genética , Conejos , Ratas
6.
J Biol Chem ; 291(48): 24838-24850, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27733688

RESUMEN

The cytokine receptor activator of NFκB ligand (RANKL) produced by osteocytes is essential for osteoclast formation in cancellous bone under physiological conditions, and RANKL production by B lymphocytes is required for the bone loss caused by estrogen deficiency. Here, we examined whether RANKL produced by osteocytes is also required for the bone loss caused by estrogen deficiency. Mice lacking RANKL in osteocytes were protected from the increase in osteoclast number and the bone loss caused by ovariectomy. Moreover, these mice did not exhibit the increase in bone marrow B lymphocytes caused by ovariectomy that occurred in control littermates. Deletion of estrogen receptor α from B cells did not alter B cell number or bone mass and did not alter the response to ovariectomy. In addition, lineage-tracing studies demonstrated that B cells do not act as osteoclast progenitors in estrogen-replete or estrogen-deficient mice. Taken together, these results demonstrate that RANKL expressed by osteocytes is required for the bone loss as well as the increase in B cell number caused by estrogen deficiency. Moreover, they suggest that estrogen control of B cell number is indirect via osteocytes and that the increase in bone marrow B cells may be a necessary component of the cascade of events that lead to cancellous bone loss during estrogen deficiency. However, the role of B cells is not to act as osteoclast progenitors but may be to act as osteoclast support cells.


Asunto(s)
Linfocitos B/metabolismo , Células de la Médula Ósea/metabolismo , Resorción Ósea/metabolismo , Estrógenos/deficiencia , Osteoclastos/metabolismo , Osteocitos/metabolismo , Ligando RANK/biosíntesis , Animales , Linfocitos B/patología , Células de la Médula Ósea/patología , Resorción Ósea/genética , Resorción Ósea/patología , Ratones , Ratones Transgénicos , Osteoclastos/patología , Osteocitos/patología , Ligando RANK/genética
7.
J Biol Chem ; 290(51): 30573-86, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26504088

RESUMEN

The biological actions of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) are mediated by the vitamin D receptor (VDR), whose expression in bone cells is regulated positively by 1,25(OH)2D3, retinoic acid, and parathyroid hormone through both intergenic and intronic enhancers. In this report, we used ChIP-sequencing analysis to confirm the presence of these Vdr gene enhancers in mesenchyme-derived bone cells and to describe the epigenetic histone landscape that spans the Vdr locus. Using bacterial artificial chromosome-minigene stable cell lines, CRISPR/Cas9 enhancer-deleted daughter cell lines, transient transfection/mutagenesis analyses, and transgenic mice, we confirmed the functionality of these bone cell enhancers in vivo as well as in vitro. We also identified VDR-binding sites across the Vdr gene locus in kidney and intestine using ChIP-sequencing analysis, revealing that only one of the bone cell-type enhancers bound VDR in kidney tissue, and none were occupied by the VDR in the intestine, consistent with weak or absent regulation by the 1,25(OH)2D3 hormone in these tissues, respectively. However, a number of additional sites of VDR binding unique to either kidney or intestine were present further upstream of the Vdr gene, suggesting the potential for alternative regulatory loci. Importantly, virtually all of these regions retained histone signatures consistent with those of enhancers and exhibited unique DNase I hypersensitivity profiles that reflected the potential for chromatin access. These studies define mechanisms associated with hormonal regulation of the Vdr and hint at the differential nature of VDR binding activity at the Vdr gene in different primary target tissues in vivo.


Asunto(s)
Calcitriol/metabolismo , Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica/fisiología , Hormonas/metabolismo , Receptores de Calcitriol/metabolismo , Animales , Calcitriol/genética , Línea Celular , Hormonas/genética , Ratones , Ratones Transgénicos , Receptores de Calcitriol/genética
8.
Am J Physiol Endocrinol Metab ; 311(3): E587-93, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27460899

RESUMEN

Glucocorticoid excess is a major cause of low bone mass and fractures. Glucocorticoid administration decreases cortical thickness and increases cortical porosity in mice, and these changes are associated with increased osteoclast number at the endocortical surface. Receptor activator of NF-κB ligand (RANKL) produced by osteocytes is required for osteoclast formation in cancellous bone as well as the increase in cortical bone resorption caused by mechanical unloading or dietary calcium deficiency. However, whether osteocyte-derived RANKL also participates in the increase in bone resorption caused by glucocorticoid excess is unknown. To address this question, we examined the effects of prednisolone on cortical bone of mice lacking RANKL production in osteocytes. Prednisolone administration increased osteoclast number at the endocortical surface, increased cortical porosity, and reduced cortical thickness in control mice, but none of these effects occurred in mice lacking RANKL in osteocytes. Prednisolone administration did not alter RANKL mRNA abundance but did reduce osteoprotegerin (OPG) mRNA abundance in osteocyte-enriched cortical bone. Similarly, dexamethasone suppressed OPG but did not increase RANKL production in cortical bone organ cultures and primary osteoblasts. These results demonstrate that RANKL produced by osteocytes is required for the cortical bone loss caused by glucocorticoid excess but suggest that the changes in endocortical resorption are driven by reduced OPG rather than elevated RANKL expression.


Asunto(s)
Resorción Ósea/genética , Glucocorticoides/farmacología , Osteocitos/metabolismo , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , Ligando RANK/biosíntesis , Animales , Resorción Ósea/inducido químicamente , Recuento de Células , Células Cultivadas , Dexametasona/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteocitos/efectos de los fármacos , Porosidad/efectos de los fármacos , Prednisolona/farmacología , Ligando RANK/genética
9.
Am J Physiol Endocrinol Metab ; 310(9): E762-73, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26956187

RESUMEN

ApoE-null (ApoE-KO) mice fed a high-fat diet (HFD) develop atherosclerosis, due in part to activation of vascular inflammation by oxidized low-density lipoprotein. Since bone loss also occurs in these mice, we used them to investigate the impact of oxidized lipids on bone homeostasis and to search for underlying pathogenic pathways. Four-month-old female ApoE-KO mice fed a HFD for three months exhibited increased levels of oxidized lipids in bone, as well as decreased femoral and vertebral trabecular and cortical bone mass, compared with ApoE-KO mice on normal diet. Despite HFD-induced increase in expression of Alox15, a lipoxygenase that oxidizes LDL and promotes atherogenesis, global deletion of this gene failed to ameliorate the skeletal impact of HFD. Osteoblast number and function were dramatically reduced in trabecular and cortical bone of HFD-fed mice, whereas osteoclast number was modestly reduced only in trabecular bone, indicating that an imbalance in favor of osteoclasts was responsible for HFD-induced bone loss. These changes were associated with decreased osteoblast progenitors and increased monocyte/macrophages in the bone marrow as well as increased expression of IL-1ß, IL-6, and TNF. HFD also attenuated Wnt signaling as evidenced by reduced expression of Wnt target genes, and it decreased expression of pro-osteoblastogenic Wnt ligands. These results suggest that oxidized lipids decrease bone mass by increasing anti-osteoblastogenic inflammatory cytokines and decreasing pro-osteoblastogenic Wnt ligands.


Asunto(s)
Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Enfermedades Óseas Metabólicas/genética , Huesos/inmunología , Osteogénesis , Proteínas Wnt/genética , Absorciometría de Fotón , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Western Blotting , Densidad Ósea , Enfermedades Óseas Metabólicas/diagnóstico por imagen , Enfermedades Óseas Metabólicas/inmunología , Enfermedades Óseas Metabólicas/patología , Huesos/diagnóstico por imagen , Huesos/metabolismo , Huesos/patología , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/inmunología , Hueso Esponjoso/metabolismo , Hueso Esponjoso/patología , Recuento de Células , Hueso Cortical/efectos de los fármacos , Hueso Cortical/inmunología , Hueso Cortical/metabolismo , Hueso Cortical/patología , Dieta Alta en Grasa , Femenino , Fémur/diagnóstico por imagen , Fémur/inmunología , Fémur/metabolismo , Fémur/patología , Citometría de Flujo , Separación Inmunomagnética , Inflamación , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipoproteínas LDL/metabolismo , Macrófagos/inmunología , Ratones , Ratones Noqueados , Monocitos/inmunología , Osteoblastos/citología , Osteoclastos/citología , Porosidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/inmunología , Columna Vertebral/metabolismo , Columna Vertebral/patología , Factor de Necrosis Tumoral alfa/genética
10.
Curr Osteoporos Rep ; 14(1): 16-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26909563

RESUMEN

The decrease in bone mass and strength during aging has multiple causes. Osteocytes are long-lived cells within the bone matrix that perform a variety of functions, including the control of bone remodeling. Because of their longevity, osteocytes are more likely than osteoclasts or osteoblasts to accumulate molecular damage over time. Osteocytes utilize quality-control pathways like autophagy to remove damaged organelles and macromolecules, and thereby maintain function. When the damage is excessive, cell death pathways such as apoptosis minimize the impact of potential osteocyte dysfunction on the skeleton. The goal of this review is to discuss how dysregulation of these pathways in osteocytes may contribute to the decline in bone mass and strength with age.


Asunto(s)
Envejecimiento/metabolismo , Apoptosis , Autofagia , Remodelación Ósea , Osteocitos/metabolismo , Osteoporosis/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Marcadores Genéticos , Humanos , Osteocitos/citología , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Transducción de Señal , Soporte de Peso
11.
J Biol Chem ; 289(35): 24069-78, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25002589

RESUMEN

A decline of the levels and activity of Sirtuin1 (Sirt1), a NAD(+) class III histone deacetylase, with age contributes to the development of several diseases including type 2 diabetes, neurodegeneration, inflammation, and cancer. The anti-aging effects of Sirt1 evidently result from the deacetylation of many transcription factors and co-factors including members of the Forkhead box O (FoxO) family and ß-catenin. Wnt/ß-catenin is indispensable for osteoblast generation. FoxOs, on the other hand, sequester ß-catenin and inhibit osteoprogenitor proliferation. Here, we have deleted Sirt1 in osteoprogenitors expressing Osterix1 (Osx1)-Cre and their descendants. Sirt1(ΔOsx1) mice had lower cortical thickness in femora and vertebrae because of reduced bone formation at the endocortical surface. In line with this, osteoprogenitor cell cultures from the Sirt1(ΔOsx1) mice exhibited lower alkaline phosphatase activity and mineralization, as well as decreased proliferation and increased apoptosis. These changes were associated with decreased Wnt/ß-catenin signaling and expression of cyclin D1 and resulted from increased binding of FoxOs to ß-catenin. These findings demonstrate that Sirt1-induced deacetylation of FoxOs unleashes Wnt signaling. A decline in Sirt1 activity in osteoblast progenitors with aging may, therefore, contribute to the age-related loss of bone mass. Together with evidence that Sirt1 activators increase bone mass in aged mice, our results also suggest that Sirt1 could be a therapeutic target for osteoporosis.


Asunto(s)
Desarrollo Óseo/fisiología , Factores de Transcripción Forkhead/fisiología , Osteoblastos/metabolismo , Sirtuina 1/fisiología , Células Madre/metabolismo , beta Catenina/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Cartilla de ADN , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sirtuina 1/genética , Proteínas Wnt/metabolismo
12.
J Biol Chem ; 288(24): 17432-40, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23645674

RESUMEN

Bone mass declines with age but the mechanisms responsible remain unclear. Here we demonstrate that deletion of a conditional allele for Atg7, a gene essential for autophagy, from osteocytes caused low bone mass in 6-month-old male and female mice. Cancellous bone volume and cortical thickness were decreased, and cortical porosity increased, in conditional knock-out mice compared with control littermates. These changes were associated with low osteoclast number, osteoblast number, bone formation rate, and wall width in the cancellous bone of conditional knock-out mice. In addition, oxidative stress was higher in the bones of conditional knock-out mice as measured by reactive oxygen species levels in the bone marrow and by p66(shc) phosphorylation in L6 vertebra. Each of these changes has been previously demonstrated in the bones of old versus young adult mice. Thus, these results demonstrate that suppression of autophagy in osteocytes mimics, in many aspects, the impact of aging on the skeleton and suggest that a decline in autophagy with age may contribute to the low bone mass associated with aging.


Asunto(s)
Fémur/metabolismo , Vértebras Lumbares/metabolismo , Osteocitos/fisiología , Envejecimiento , Animales , Autofagia , Proteína 7 Relacionada con la Autofagia , Densidad Ósea , Diferenciación Celular , Células Cultivadas , Femenino , Fémur/diagnóstico por imagen , Fémur/patología , Vértebras Lumbares/diagnóstico por imagen , Vértebras Lumbares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Osteoblastos/fisiología , Osteoclastos/fisiología , Estrés Oxidativo , Radiografía , Especies Reactivas de Oxígeno/metabolismo
13.
Bone ; : 117181, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38960295

RESUMEN

Staphylococcus aureus osteomyelitis leads to extensive bone destruction. Osteoclasts are bone resorbing cells that are often increased in bone infected with S. aureus. The cytokine RANKL is essential for osteoclast formation under physiological conditions but in vitro evidence suggests that inflammatory cytokines may by-pass the requirement for RANKL. The goal of this study was to determine whether RANKL-dependent osteoclast formation is essential for the bone loss that occurs in a murine model of S. aureus osteomyelitis. To this end, humanized-RANKL mice were infected by direct inoculation of S. aureus into a unicortical defect in the femur. Mice were treated with vehicle or denosumab, a human monoclonal antibody that inhibits RANKL, both before and during a 14-day infection period. The severe cortical bone destruction caused by infection was completely prevented by denosumab administration even though the bacterial burden in the femur was not affected. Osteoclasts were abundant near the inoculation site in vehicle-treated mice but absent in denosumab-treated mice. In situ hybridization demonstrated that S. aureus infection potently stimulated RANKL expression in bone marrow stromal cells. The extensive reactive bone formation that occurs in this osteomyelitis model was also reduced by denosumab administration. Lastly, there was a notable lack of osteoblasts near the infection site suggesting that the normal coupling of bone formation to bone resorption was disrupted by S. aureus infection. These results demonstrate that RANKL-mediated osteoclast formation is required for the bone loss that occurs in S. aureus infection and suggest that disruption of the coupling of bone formation to bone resorption may also contribute to bone loss in this condition.

14.
Res Sq ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38558984

RESUMEN

Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression that disrupts the balance between osteoclasts and osteoblasts, leading to bone lesions. Whether such reprogramming affects matrix-embedded osteocytes remains poorly understood. Here, we demonstrate that osteocytes in breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified osteocytes from mice with breast cancer bone metastasis enriched in senescence and SASP markers and pro-osteoclastogenic genes. Using multiplex in situ hybridization and AI-assisted analysis, we detected osteocytes with senescence-associated distension of satellites, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. In vitro and ex vivo organ cultures showed that breast cancer cells promote osteocyte senescence and enhance their osteoclastogenic potential. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that osteocytes undergo pathological reprogramming by breast cancer cells and identify osteocyte senescence as an initiating event triggering bone destruction in breast cancer metastases.

15.
J Biol Chem ; 287(35): 29851-60, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22782898

RESUMEN

Production of the cytokine receptor activator of NFκB ligand (RANKL) by lymphocytes has been proposed as a mechanism by which sex steroid deficiency causes bone loss. However, there have been no studies that functionally link RANKL expression in lymphocytes with bone loss in this condition. Herein, we examined whether RANKL expression in either B or T lymphocytes contributes to ovariectomy-induced bone loss in mice. Mice harboring a conditional RANKL allele were crossed with CD19-Cre or Lck-Cre mice to delete RANKL in B or T lymphocytes, respectively. Deletion of RANKL from either cell type had no impact on bone mass in estrogen-replete mice up to 7 months of age. However, mice lacking RANKL in B lymphocytes were partially protected from the bone loss caused by ovariectomy. This protection occurred in cancellous, but not cortical, bone and was associated with a failure to increase osteoclast numbers in the conditional knock-out mice. Deletion of RANKL from T lymphocytes had no impact on ovariectomy-induced bone loss. These results demonstrate that lymphocyte RANKL is not involved in basal bone remodeling, but B cell RANKL does contribute to the increase in osteoclasts and cancellous bone loss that occurs after loss of estrogen.


Asunto(s)
Linfocitos B/metabolismo , Remodelación Ósea/inmunología , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Ligando RANK/metabolismo , Alelos , Animales , Linfocitos B/inmunología , Densidad Ósea/genética , Densidad Ósea/inmunología , Estrógenos/genética , Estrógenos/inmunología , Estrógenos/metabolismo , Femenino , Humanos , Ratones , Ratones Transgénicos , Osteoclastos/inmunología , Osteoporosis/genética , Osteoporosis/inmunología , Ovariectomía , Ligando RANK/genética , Ligando RANK/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Annu Rev Pathol ; 18: 257-281, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36207010

RESUMEN

Osteoclasts are multinucleated cells with the unique ability to resorb bone matrix. Excessive production or activation of osteoclasts leads to skeletal pathologies that affect a significant portion of the population. Although therapies that effectively target osteoclasts have been developed, they are associated with sometimes severe side effects, and a fuller understanding of osteoclast biology may lead to more specific treatments. Along those lines, a rich body of work has defined essential signaling pathways required for osteoclast formation, function, and survival. Nonetheless, recent studies have cast new light on long-held views regarding the origin of these cells during development and homeostasis, their life span, and the cellular sources of factors that drive their production and activity during homeostasis and disease. In this review, we discuss these new findings in the context of existing work and highlight areas of ongoing and future investigation.


Asunto(s)
Resorción Ósea , Osteoclastos , Humanos , Osteoclastos/metabolismo , Osteoclastos/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Resorción Ósea/patología , Transducción de Señal/fisiología , Diferenciación Celular
17.
Bone ; 170: 116702, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36773885

RESUMEN

The scavenger receptor class B member 1 (SR-B1 or Scarb1) is a glycosylated cell surface receptor for high density lipoproteins (HDL), oxidized low density lipoproteins (OxLDL), and phosphocholine-containing oxidized phospholipids (PC-OxPLs). Scarb1 is expressed in macrophages and has been shown to have both pro- and anti-atherogenic properties. It has been reported that global deletion of Scarb1 in mice leads to either high or low bone mass and that PC-OxPLs decrease osteoblastogenesis and increase osteoclastogenesis. PC-OxPLs decrease bone mass in 6-month-old mice and are critical pathogenetic factors in the bone loss caused by high fat diet or aging. We have investigated here whether Scarb1 expression in myeloid cells affects bone mass and whether PC-OxPLs exert their anti-osteogenic effects via activation of Scarb1 in macrophages. To this end, we generated mice with deletion of Scarb1 in LysM-Cre expressing cells and found that lack of Scarb1 did not affect bone mass in vivo. These results indicate that Scarb1 expression in cells of the myeloid/osteoclast lineage does not contribute to bone homeostasis. Based on this evidence, and earlier studies of ours showing that Scarb1 expression in osteoblasts does not affect bone mass, we conclude that Scarb1 is not an important mediator of the adverse effects on PC-OxPLs in osteoclasts or osteoblasts in 6-month-old mice.


Asunto(s)
Densidad Ósea , Huesos , Animales , Ratones , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Huesos/metabolismo , Osteoclastos/metabolismo , Osteogénesis
18.
bioRxiv ; 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38014179

RESUMEN

Single-cell RNA sequencing has led to numerous novel designations for mesenchymal cell types associated with bone. Consequently, there are now multiple designations for what appear to be the same cell type. In addition, existing datasets contain relatively small numbers of mature osteoblasts and osteocytes and there has been no comparison of periosteal bone cells to those at the endosteum and trabecular bone. The main goals of this study were to increase the amount of single cell RNA sequence data for osteoblasts and osteocytes, to compare cells from the periosteum to those inside bone, and to clarify the major categories of cell types associated with murine bone. To do this, we created an atlas of murine bone-associated cells by harmonizing published datasets with in-house data from cells targeted by Osx1-Cre and Dmp1-Cre driver strains. Cells from periosteal bone were analyzed separately from those isolated from the endosteum and trabecular bone. Over 100,000 mesenchymal cells were mapped to reveal 11 major clusters designated fibro-1, fibro-2, chondrocytes, articular chondrocytes, tenocytes, adipo-CAR, osteo-CAR, pre-osteoblasts, osteoblasts, osteocytes, and osteo-X, the latter defined in part by Postn expression. Osteo-X, osteo-CAR, and pre-osteoblasts were closely associated with osteoblasts at the trabecular bone surface. Wnt16 was expressed in multiple cell types from the periosteum but not in any cells from endocortical or cancellous bone. Fibro-2 cells, which express markers of skeletal stem cells, localized to the periosteum but not trabecular bone in adult mice. Suppressing bone remodeling eliminated osteoblasts and altered gene expression in pre-osteoblasts but did not change the abundance or location of osteo-X or osteo-CAR cells. These results provide a framework for identifying bone cell types in murine single cell RNA sequencing datasets and suggest that osteoblast progenitors reside near the surface of remodeling bone.

19.
JCI Insight ; 8(18)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37581932

RESUMEN

Denosumab is an anti-RANKL Ab that potently suppresses bone resorption, increases bone mass, and reduces fracture risk. Discontinuation of denosumab causes rapid rebound bone resorption and bone loss, but the molecular mechanisms are unclear. We generated humanized RANKL mice and treated them with denosumab to examine the cellular and molecular conditions associated with rebound resorption. Denosumab potently suppressed both osteoclast and osteoblast numbers in cancellous bone in humanized RANKL mice. The decrease in osteoclast number was not associated with changes in osteoclast progenitors in bone marrow. Long-term, but not short-term, denosumab administration reduced osteoprotegerin (OPG) mRNA in bone. Localization of OPG expression revealed that OPG mRNA is produced by a subpopulation of osteocytes. Long-term denosumab administration reduced osteocyte OPG mRNA, suggesting that OPG expression declines as osteocytes age. Consistent with this, osteocyte expression of OPG was more prevalent near the surface of cortical bone in humans and mice. These results suggest that new osteocytes are an important source of OPG in remodeling bone and that suppression of remodeling reduces OPG abundance by reducing new osteocyte formation. The lack of new osteocytes and the OPG they produce may contribute to rebound resorption after denosumab discontinuation.


Asunto(s)
Resorción Ósea , Osteocitos , Humanos , Ratones , Animales , Osteocitos/metabolismo , Denosumab/farmacología , Denosumab/uso terapéutico , Denosumab/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Osteoclastos/metabolismo , Resorción Ósea/metabolismo
20.
Nat Commun ; 14(1): 3616, 2023 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-37330524

RESUMEN

NAD is an essential co-factor for cellular energy metabolism and multiple other processes. Systemic NAD+ deficiency has been implicated in skeletal deformities during development in both humans and mice. NAD levels are maintained by multiple synthetic pathways but which ones are important in bone forming cells is unknown. Here, we generate mice with deletion of Nicotinamide Phosphoribosyltransferase (Nampt), a critical enzyme in the NAD salvage pathway, in all mesenchymal lineage cells of the limbs. At birth, NamptΔPrx1 exhibit dramatic limb shortening due to death of growth plate chondrocytes. Administration of the NAD precursor nicotinamide riboside during pregnancy prevents the majority of in utero defects. Depletion of NAD post-birth also promotes chondrocyte death, preventing further endochondral ossification and joint development. In contrast, osteoblast formation still occurs in knockout mice, in line with distinctly different microenvironments and reliance on redox reactions between chondrocytes and osteoblasts. These findings define a critical role for cell-autonomous NAD homeostasis during endochondral bone formation.


Asunto(s)
Metabolismo Energético , NAD , Humanos , Ratones , Animales , NAD/metabolismo , Oxidación-Reducción , Homeostasis , Ratones Noqueados , Citocinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA