Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Calcif Tissue Int ; 113(1): 96-109, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37243756

RESUMO

Differentiation and optimal function of osteoblasts and osteoclasts are contingent on synthesis and maintenance of a healthy proteome. Impaired and/or altered secretory capacity of these skeletal cells is a primary driver of most skeletal diseases. The endoplasmic reticulum (ER) orchestrates the folding and maturation of membrane as well as secreted proteins at high rates within a calcium rich and oxidative organellar niche. Three ER membrane proteins monitor fidelity of protein processing in the ER and initiate an intricate signaling cascade known as the Unfolded Protein Response (UPR) to remediate accumulation of misfolded proteins in its lumen, a condition referred to as ER stress. The UPR aids in fine-tuning, expanding and/or modifying  the cellular proteome, especially in specialized secretory cells, to match everchanging physiologic cues and metabolic demands. Sustained activation of the UPR due to chronic ER stress, however, is known to hasten cell death and drive pathophysiology of several diseases. A growing body of evidence suggests that ER stress and an aberrant UPR may contribute to poor skeletal health and the development of osteoporosis. Small molecule therapeutics that target distinct components of the UPR may therefore have implications for developing novel treatment modalities relevant to the skeleton. This review summarizes the complexity of UPR actions in bone cells in the context of skeletal physiology and osteoporotic bone loss, and highlights the need for future mechanistic studies to develop novel UPR therapeutics that mitigate adverse skeletal outcomes.


Assuntos
Proteoma , Resposta a Proteínas não Dobradas , Proteoma/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Transdução de Sinais , Retículo Endoplasmático/metabolismo
2.
J Biol Chem ; 289(35): 24069-78, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25002589

RESUMO

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.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Osteoblastos/metabolismo , Sirtuína 1/fisiologia , Células-Tronco/metabolismo , beta Catenina/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Proliferação de Células , Primers do DNA , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Sirtuína 1/genética , Proteínas Wnt/metabolismo
3.
Sci Rep ; 12(1): 10257, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715555

RESUMO

The protective effect of estrogens against cortical bone loss is mediated via direct actions on mesenchymal cells, but functional evidence for the mediators of these effects has only recently begun to emerge. We report that the matrix metalloproteinase 13 (MMP13) is the highest up-regulated gene in mesenchymal cells from mice lacking the estrogen receptor alpha (ERα). In sham-operated female mice with conditional Mmp13 deletion in Prrx1 expressing cells (Mmp13ΔPrrx1), the femur and tibia length was lower as compared to control littermates (Mmp13f./f). Additionally, in the sham-operated female Mmp13ΔPrrx1 mice cortical thickness and trabecular bone volume in the femur and tibia were higher and osteoclast number at the endocortical surfaces was lower, whereas bone formation rate was unaffected. Notably, the decrease of cortical thickness caused by ovariectomy (OVX) in the femur and tibia of Mmp13f./f mice was attenuated in the Mmp13ΔPrrx1 mice; but the decrease of trabecular bone caused by OVX was not affected. These results reveal that mesenchymal cell-derived MMP13 may regulate osteoclast number and/or activity, bone resorption, and bone mass. And increased production of mesenchymal cell-derived factors may be important mediators of the adverse effect of estrogen deficiency on cortical, but not trabecular, bone.


Assuntos
Densidade Óssea , Doenças Ósseas Metabólicas , Metaloproteinase 13 da Matriz/metabolismo , Animais , Osso e Ossos/diagnóstico por imagem , Osso Cortical , Estrogênios , Feminino , Proteínas de Homeodomínio , Humanos , Metaloproteinase 13 da Matriz/genética , Camundongos , Ovariectomia/efeitos adversos
4.
Bioessays ; 31(2): 209-19, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19204993

RESUMO

Studies on Neurospora chromosome segment duplications (Dps) performed since the publication of Perkins's comprehensive review in 1997 form the focus of this article. We present a brief summary of Perkins's seminal work on chromosome rearrangements, specifically, the identification of insertional and quasiterminal translocations that can segregate Dp progeny when crossed with normal sequence strains (i.e., T x N). We describe the genome defense process called meiotic silencing by unpaired DNA that renders Dp-heterozygous crosses (i.e., Dp x N) barren, which provides a basis for identifying Dps, and discuss whether other processes also might contribute to the barren phenotype of Dp x N and Dp x Dp crosses. We then turn to studies suggesting that large Dps (i.e., >300 kbp) can allow smaller gene-sized duplications to escape another genome defense process called repeat-induced point mutation (RIP), possibly by titration of the RIP machinery. Finally, we assess whether in natural populations dominant RIP suppressor Dps provide an "RIP-free" niche for evolution of new genes following the duplication of existing genes.


Assuntos
Cromossomos Fúngicos/genética , Cruzamentos Genéticos , Duplicação Gênica , Neurospora crassa/genética , Heterozigoto , Meiose , Fenótipo
5.
NPJ Aging Mech Dis ; 7(1): 8, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33795658

RESUMO

Age-related osteoporosis is caused by a deficit in osteoblasts, the cells that secrete bone matrix. The number of osteoblast progenitors also declines with age associated with increased markers of cell senescence. The forkhead box O (FoxO) transcription factors attenuate Wnt/ß-catenin signaling and the proliferation of osteoprogenitors, thereby decreasing bone formation. The NAD+-dependent Sirtuin1 (Sirt1) deacetylates FoxOs and ß-catenin in osteoblast progenitors and, thereby, increases bone mass. However, it remains unknown whether the Sirt1/FoxO/ß-catenin pathway is dysregulated with age in osteoblast progenitors. We found decreased levels of NAD+ in osteoblast progenitor cultures from old mice, associated with increased acetylation of FoxO1 and markers of cell senescence. The NAD+ precursor nicotinamide riboside (NR) abrogated FoxO1 and ß-catenin acetylation and several marker of cellular senescence, and increased the osteoblastogenic capacity of cells from old mice. Consistent with these effects, NR administration to C57BL/6 mice counteracted the loss of bone mass with aging. Attenuation of NAD+ levels in osteoprogenitor cultures from young mice inhibited osteoblastogenesis in a FoxO-dependent manner. In addition, mice with decreased NAD+ in cells of the osteoblast lineage lost bone mass at a young age. Together, these findings suggest that the decrease in bone formation with old age is due, at least in part, to a decrease in NAD+ and dysregulated Sirt1/FoxO/ß-catenin pathway in osteoblast progenitors. NAD+ repletion, therefore, represents a rational therapeutic approach to skeletal involution.

6.
J Bone Miner Res ; 35(8): 1441-1451, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32154948

RESUMO

CXCL12 is abundantly expressed in reticular cells associated with the perivascular niches of the bone marrow (BM) and is indispensable for B lymphopoiesis. Cxcl12 promotes osteoclastogenesis and has been implicated in pathologic bone resorption. We had shown earlier that estrogen receptor α deletion in osteoprogenitors and estrogen deficiency in mice increase Cxcl12 mRNA and protein levels in the BM plasma, respectively. We have now generated female and male mice with conditional deletion of a Cxcl12 allele in Prrx1 targeted cells (Cxcl12∆Prrx1 ) and show herein that they have a 90% decrease in B lymphocytes but increased erythrocytes and adipocytes in the marrow. Ovariectomy increased the expression of Cxcl12 and B-cell number in the Cxcl12f/f control mice, but these effects were abrogated in the Cxcl12∆Prrx1 mice. Cortical bone mass was not affected in Cxcl12∆Prrx1 mice. Albeit, the cortical bone loss caused by ovariectomy was greatly attenuated. Most unexpectedly, the rate of bone turnover in sex steroid-sufficient female or male Cxcl12∆Prrx1 mice was dramatically increased, as evidenced by a more than twofold increase in several osteoblast- and osteoclast-specific mRNAs, as well as increased mineral apposition and bone formation rate and increased osteoclast number in the endosteal surface. The magnitude of the Cxcl12∆Prrx1 -induced changes were much greater than those caused by ovariectomy or orchidectomy in the Cxcl12f/f mice. These results strengthen the evidence that CXCL12 contributes to the loss of cortical bone mass caused by estrogen deficiency. Moreover, they reveal for the first time that in addition to its effects on hematopoiesis, CXCL12 restrains bone turnover-without changing the balance between resorption and formation-by suppressing osteoblastogenesis and the osteoclastogenesis support provided by cells of the osteoblast lineage. © 2020 American Society for Bone and Mineral Research.


Assuntos
Reabsorção Óssea , Osso e Ossos , Quimiocina CXCL12 , Estrogênios/deficiência , Animais , Remodelação Óssea , Reabsorção Óssea/genética , Quimiocina CXCL12/genética , Osso Cortical/diagnóstico por imagem , Feminino , Deleção de Genes , Proteínas de Homeodomínio , Masculino , Camundongos , Osteoblastos , Osteoclastos , Ovariectomia
7.
FASEB Bioadv ; 2(4): 207-218, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32259048

RESUMO

Increased production of the osteoclastogenic cytokine RANKL is a common feature of pathologic bone loss, but the underlying cause of this increase is poorly understood. The unfolded protein response (UPR) is activated in response to accumulation of misfolded proteins in the endoplasmic reticulum (ER). Failure to resolve misfolding results in excess UPR signaling that stimulates cytokine production and cell death. We therefore investigated whether RANKL is one of the cytokines stimulated in response to elevated UPR in bone cells. Pharmacologic induction of UPR with tunicamycin (Tm)-stimulated RANKL expression in cultures of primary osteoblastic cells and in osteoblast and osteocyte cell lines. Pharmacologic inhibition of the UPR blunted Tm-induced RANKL production. Silencing Edem1 or Sel1l, proteins that aid in degradation of misfolded proteins, also induced UPR and increased RANKL mRNA. Moreover, Tm or hypoxia increased RANKL and bone resorption in cultures of neonatal murine calvaria. Administration of Tm to adult mice caused dilation of ER in osteoblasts and osteocytes, elevated the UPR, and increased RANKL expression and osteoclast number. These findings support the hypothesis that excessive UPR signaling stimulates the expression of RANKL by osteoblasts and osteocytes, and thereby facilitates excessive bone resorption and bone loss in pathologic conditions.

8.
Cell Death Discov ; 6: 8, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32123584

RESUMO

Genetic obesity increases in liver phosphatidylcholine (PC)/phosphatidylethanolamine (PE) ratio, inducing endoplasmic reticulum (ER) stress without concomitant increase of ER chaperones. Here, it is found that exposing mice to a palm oil-based high fat (HF) diet induced obesity, loss of liver PE, and loss of the ER chaperone Grp78/BiP in pericentral hepatocytes. In Hepa1-6 cells treated with elevated concentration of palmitate to model lipid stress, Grp78/BiP mRNA was increased, indicating onset of stress-induced Unfolded Protein Response (UPR), but Grp78/BiP protein abundance was nevertheless decreased. Exposure to elevated palmitate also induced in hepatoma cells decreased membrane glycosylation, nuclear translocation of pro-apoptotic C/EBP-homologous-protein-10 (CHOP), expansion of ER-derived quality control compartment (ERQC), loss of mitochondrial membrane potential (MMP), and decreased oxidative phosphorylation. When PE was delivered to Hepa1-6 cells exposed to elevated palmitate, effects by elevated palmitate to decrease Grp78/BiP protein abundance and suppress membrane glycosylation were blunted. Delivery of PE to Hepa1-6 cells treated with elevated palmitate also blunted expansion of ERQC, decreased nuclear translocation of CHOP and lowered abundance of reactive oxygen species (ROS). Instead, delivery of the chemical chaperone 4-phenyl-butyrate (PBA) to Hepa1-6 cells treated with elevated palmitate, while increasing abundance of Grp78/BiP protein and restoring membrane glycosylation, also increased ERQC, expression and nuclear translocation of CHOP, non-mitochondrial oxygen consumption, and generation of ROS. Data indicate that delivery of PE to hepatoma cells under lipid stress recovers cell function by targeting the secretory pathway and by blunting pro-apoptotic branches of the UPR.

9.
JCI Insight ; 5(19)2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32870816

RESUMO

In aging mice, osteoclast number increases in cortical bone but declines in trabecular bone, suggesting that different mechanisms underlie age-associated bone loss in these 2 compartments. Osteocytes produce the osteoclastogenic cytokine RANKL, encoded by Tnfsf11. Tnfsf11 mRNA increases in cortical bone of aged mice, suggesting a mechanism underlying the bone loss. To address this possibility, we aged mice lacking RANKL in osteocytes. Whereas control mice lost cortical bone between 8 and 24 months of age, mice lacking RANKL in osteocytes gained cortical bone during this period. Mice of both genotypes lost trabecular bone with age. Osteoclasts increased with age in cortical bone of control mice but not in RANKL conditional knockout mice. Induction of cellular senescence increased RANKL production in murine and human cell culture models, suggesting an explanation for elevated RANKL levels with age. Overexpression of the senescence-associated transcription factor Gata4 stimulated Tnfsf11 expression in cultured murine osteoblastic cells. Finally, elimination of senescent cells from aged mice using senolytic compounds reduced Tnfsf11 mRNA in cortical bone. Our results demonstrate the requirement of osteocyte-derived RANKL for age-associated cortical bone loss and suggest that increased Tnfsf11 expression with age results from accumulation of senescent cells in cortical bone.


Assuntos
Envelhecimento/patologia , Reabsorção Óssea/patologia , Senescência Celular , Osso Cortical/patologia , Osteócitos/patologia , Ligante RANK/fisiologia , Envelhecimento/metabolismo , Animais , Reabsorção Óssea/etiologia , Reabsorção Óssea/metabolismo , Osso Cortical/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteócitos/metabolismo
10.
Sci Rep ; 10(1): 11933, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32686739

RESUMO

Loss of estrogens at menopause is a major cause of osteoporosis and increased fracture risk. Estrogens protect against bone loss by decreasing osteoclast number through direct actions on cells of the myeloid lineage. Here, we investigated the molecular mechanism of this effect. We report that 17ß-estradiol (E2) decreased osteoclast number by promoting the apoptosis of early osteoclast progenitors, but not mature osteoclasts. This effect was abrogated in cells lacking Bak/Bax-two pro-apoptotic members of the Bcl-2 family of proteins required for mitochondrial apoptotic death. FasL has been previously implicated in the pro-apoptotic actions of E2. However, we show herein that FasL-deficient mice lose bone mass following ovariectomy indistinguishably from FasL-intact controls, indicating that FasL is not a major contributor to the anti-osteoclastogenic actions of estrogens. Instead, using microarray analysis we have elucidated that ERα-mediated estrogen signaling in osteoclast progenitors decreases "oxidative phosphorylation" and the expression of mitochondria complex I genes. Additionally, E2 decreased the activity of complex I and oxygen consumption rate. Similar to E2, the complex I inhibitor Rotenone decreased osteoclastogenesis by promoting osteoclast progenitor apoptosis via Bak/Bax. These findings demonstrate that estrogens decrease osteoclast number by attenuating respiration, and thereby, promoting mitochondrial apoptotic death of early osteoclast progenitors.


Assuntos
Trifosfato de Adenosina/biossíntese , Estrogênios/metabolismo , Mitocôndrias/metabolismo , Células Precursoras de Monócitos e Macrófagos/metabolismo , Osteoclastos/metabolismo , Fosforilação Oxidativa , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Densidade Óssea , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Contagem de Células , Diferenciação Celular , Células Cultivadas , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais
11.
Aging Cell ; 18(3): e12923, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30773784

RESUMO

Both an increase in osteoclast and a decrease in osteoblast numbers contribute to skeletal aging. Markers of cellular senescence, including expression of the cyclin inhibitor p16, increase with aging in several bone cell populations. The elimination of p16-expressing cells in old mice, using the INK-ATTAC transgene, increases bone mass indicating that senescent cells contribute to skeletal aging. However, the identity of the senescent cells and the extent to which ablation of p16-expressing cells may prevent skeletal aging remain unknown. Using mice expressing the p16-3MR transgene, we examined whether elimination of p16-expressing cells between 12 and 24 months of age could preserve bone mass; and whether elimination of these cells from 20 to 26 months of age could restore bone mass. The activation of the p16-3MR transgene by ganciclovir (GCV) greatly diminished p16 levels in the brain, liver, and osteoclast progenitors from the bone marrow. The age-related increase in osteoclastogenic potential of myeloid cells was also abrogated by GCV. However, GCV did not alter p16 levels in osteocytes-the most abundant cell type in bone-and had no effect on the skeletal aging of p16-3MR mice. These findings indicate that the p16-3MR transgene does not eliminate senescent osteocytes but it does eliminate senescent osteoclast progenitors and senescent cells in other tissues, as described previously. Elimination of senescent osteoclast progenitors, in and of itself, has no effect on the age-related loss of bone mass. Hence, other senescent cell types, such as osteocytes, must be the seminal culprits.


Assuntos
Envelhecimento , Densidade Óssea , Senescência Celular , Osteoclastos/citologia , Células-Tronco/citologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
12.
Curr Top Dev Biol ; 127: 149-163, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29433736

RESUMO

Recent studies with murine models of cell-specific loss- or gain-of-function of FoxOs have provided novel insights into the function and signaling of these transcription factors on the skeleton. They have revealed that FoxO actions in chondrocytes are critical for normal skeletal development, and FoxO actions in cells of the osteoclast or osteoblast lineage greatly influence bone resorption and formation and, consequently, bone mass. FoxOs also act in osteoblast progenitors to inhibit Wnt signaling and bone formation. Additionally, FoxOs decrease bone resorption via direct antioxidant effects on osteoclasts and upregulation of the antiosteoclastogenic cytokine OPG in cells of the osteoblast lineage. Deacetylation of FoxOs by the NAD-dependent histone deacetylase Sirt1 in both osteoblasts and osteoclasts stimulates bone formation and inhibits bone resorption, making Sirt1 activators promising therapeutic agents for diseases of low bone mass. In this chapter, we review these advances and discuss their implications for the pathogenesis and treatment of estrogen deficiency-, Type 1 diabetes-, and age-related osteoporosis.


Assuntos
Reabsorção Óssea/metabolismo , Osso e Ossos/metabolismo , Osteogênese , Animais , Reabsorção Óssea/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Mutação , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transdução de Sinais/genética
13.
Dent Res J (Isfahan) ; 14(5): 326-330, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29109747

RESUMO

BACKGROUND: Surface roughness associated with improper finishing/polishing of restorations can result in plaque accumulation, gingival irritation, surface staining, and poor esthetic of restored teeth. The study aimed to evaluate the efficiency of various finishing and polishing systems and time using various procedures on surface roughness of some esthetic restorative materials. MATERIALS AND METHODS: In this in vitro study, samples of two composite materials, compomer and glass ionomer cement (GIC) materials, were fabricated. Finishing and polishing were done immediately (n = 40) and after 1 week (n = 40) using four systems (diamond bur + soflex discs; diamond bur + Astropol polishing brush; tungsten carbide bur + soflex discs; tungsten carbide bur + Astropol polishing brush). Surface roughness was measured using surface profilometer. Data were statistically analyzed by t-test (for each material and time period) and one-way analysis of variance followed by Tukey's post hoc (for finishing and polishing systems) at a significant level of P < 0.05. RESULTS: Analysis of time period, irrespective of finishing and polishing system showed that Ra values were greater (P < 0.05) in delayed polishing in GIC > Z100 > Filtek P90 > Dyract AP, suggesting immediate polishing is better. Among the materials, Filtek P90 had the least Ra values indicating the smoothest surface among all materials, followed by Z100, Dyract AP, and GIC. Comparison of polishing and finishing systems irrespective of materials showed that Ra values were lower (P > 0.05) in diamond + Astropol combination whereas diamond + soflex had the greatest Ra values. CONCLUSION: It might be concluded that: (i) Filtek P90 showed least Ra values followed by < Z100 < Dyract < GIC; (ii) immediate (24 h) finishing/polishing of materials is better than delayed; and (iii) among all these polishing systems, diamond bur-Astropol and Astrobrush showed good surface finish.

14.
J Bone Miner Res ; 32(1): 60-69, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27491024

RESUMO

Type 1 diabetes is associated with osteopenia and increased fragility fractures, attributed to reduced bone formation. However, the molecular mechanisms mediating these effects remain unknown. Insulin promotes osteoblast formation and inhibits the activity of the FoxO transcription factors. FoxOs, on the other hand, inhibit osteoprogenitor proliferation and bone formation. Here, we investigated whether FoxOs play a role in the low bone mass associated with type 1 diabetes, using mice lacking FoxO1, 3, and 4 in osteoprogenitor cells (FoxO1,3,4ΔOsx1-Cre ). Streptozotocin-induced diabetes caused a reduction in bone mass and strength in FoxO-intact mice. In contrast, cancellous bone was unaffected in diabetic FoxO1,3,4ΔOsx1-Cre mice. The low bone mass in the FoxO-intact diabetic mice was associated with decreased osteoblast number and bone formation, as well as decreased expression of the anti-osteoclastogenic cytokine osteoprotegerin (OPG) and increased osteoclast number. FoxO deficiency did not alter the effects of diabetes on bone formation; however, it did prevent the decrease in OPG and the increase in osteoclast number. Addition of high glucose to osteoblastic cell cultures decreased OPG mRNA, indicating that hyperglycemia in and of itself contributes to diabetic bone loss. Taken together, these results suggest that FoxOs exacerbate the loss of cancellous bone mass associated with type 1 diabetes and that inactivation of FoxOs might ameliorate the adverse effects of insulin deficiency. © 2016 American Society for Bone and Mineral Research.


Assuntos
Osso Esponjoso/patologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Fatores de Transcrição Forkhead/metabolismo , Deleção de Genes , Osteoblastos/metabolismo , Células-Tronco/metabolismo , Animais , Reabsorção Óssea/complicações , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/patologia , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/metabolismo , Linhagem da Célula , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 1/complicações , Modelos Animais de Doenças , Feminino , Fêmur/diagnóstico por imagem , Fêmur/patologia , Glucose/metabolismo , Integrases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Osteoblastos/patologia , Osteoprotegerina/metabolismo , Microtomografia por Raio-X
15.
Aging Cell ; 16(4): 693-703, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28401730

RESUMO

Age-related bone loss in mice results from a decrease in bone formation and an increase in cortical bone resorption. The former is accounted by a decrease in the number of postmitotic osteoblasts which synthesize the bone matrix and is thought to be the consequence of age-dependent changes in mesenchymal osteoblast progenitors. However, there are no specific markers for these progenitors, and conclusions rely on results from in vitro cultures of mixed cell populations. Moreover, the culprits of such changes remain unknown. Here, we have used Osx1-Cre;TdRFP mice in which osteoprogenitors express the TdRFP fluorescent protein. We report that the number of TdRFP-Osx1 cells, freshly isolated from the bone marrow, declines by more than 50% between 6 and 24 months of age in both female and male mice. Moreover, TdRFP-Osx1 cells from old mice exhibited markers of DNA damage and senescence, such as γH2AX foci, G1 cell cycle arrest, phosphorylation of p53, increased p21CIP1 levels, as well as increased levels of GATA4 and activation of NF-κB - two major stimulators of the senescence-associated secretory phenotype (SASP). Bone marrow stromal cells from old mice also exhibited elevated expression of SASP genes, including several pro-osteoclastogenic cytokines, and increased capacity to support osteoclast formation. These changes were greatly attenuated by the senolytic drug ABT263. Together, these findings suggest that the decline in bone mass with age is the result of intrinsic defects in osteoprogenitor cells, leading to decreased osteoblast numbers and increased support of osteoclast formation.


Assuntos
Envelhecimento/genética , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Osteoporose/genética , Fator de Transcrição Sp7/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Compostos de Anilina/farmacologia , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Histonas/genética , Histonas/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteoporose/metabolismo , Osteoporose/patologia , Cultura Primária de Células , Transdução de Sinais , Fator de Transcrição Sp7/metabolismo , Sulfonamidas/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Vermelha Fluorescente
16.
J Bone Miner Res ; 32(3): 560-574, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27714847

RESUMO

Old age and sex steroid deficiency are the two most critical factors for the development of osteoporosis. It remains unknown, however, whether the molecular culprits of the two conditions are similar or distinct. We show herein that at 19.5 months of age-a time by which the age-dependent decline of cortical and cancellous bone mass and cortical porosity were fully manifested in C57BL/6J mice-these animals remained functionally estrogen sufficient. Transgenic mice with conditional expression of mitochondria-targeted catalase-a potent H2 O2 inactivating enzyme-in cells of the myeloid lineage (mitoCAT;LysM-Cre mice) were protected from the loss of cortical, but not cancellous, bone caused by gonadectomy in either sex. Consistent with these findings, in vitro studies with ERα-deficient Prx1+ cells and gonadectomized young adult mice showed that in both sexes decreased ERα signaling in Prx1+ cells leads to an increase in SDF1, a.k.a. CXCL12, an osteoclastogenic cytokine whose effects were abrogated in macrophages from mitoCAT;LysM-Cre mice. In contrast to sex steroid deficiency, the adverse effects of aging on either cortical or cancellous bone were unaffected in mitoCAT;LysM-Cre mice. On the other hand, attenuation of H2 O2 generation in cells of the mesenchymal lineage targeted by Prx1-Cre partially prevented the loss of cortical bone caused by old age. Our results suggest the effects of sex steroid deficiency and aging on the murine skeleton are independent and result from distinct mechanisms. In the former, the prevailing mechanism of the cortical bone loss in both sexes is increased osteoclastogenesis caused by estrogen deficiency; this is likely driven, at least in part, by mesenchymal/stromal cell-derived SDF1. Decreased osteoblastogenesis, owing in part to increased H2 O2, combined with increased osteoclastogenesis caused by aging mechanisms independent of estrogen deficiency, are the prevailing mechanisms of the loss of cortical bone with old age. © 2016 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento/fisiologia , Osso e Ossos/fisiologia , Hormônios Esteroides Gonadais/deficiência , Animais , Fenômenos Biomecânicos , Reabsorção Óssea/patologia , Reabsorção Óssea/fisiopatologia , Calcificação Fisiológica , Osso Esponjoso/fisiologia , Contagem de Células , Linhagem da Célula , Quimiocina CXCL12/metabolismo , Osso Cortical/fisiologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Hormônios Esteroides Gonadais/metabolismo , Peróxido de Hidrogênio/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Osteoclastos/metabolismo , Ovariectomia , Porosidade
17.
Mol Endocrinol ; 29(10): 1498-509, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26287518

RESUMO

Activation of Sirtuin1 (Sirt1), an nicotinamide adenine dinucleotide oxidized-dependent deacetylase, by natural or synthetic compounds like resveratrol, SRT2104, or SRT3025 attenuates the loss of bone mass caused by ovariectomy, aging, or unloading in mice. Conversely, Sirt1 deletion in osteoclast progenitors increases osteoclast number and bone resorption. Sirt1 deacetylates forkhead box protein (Fox) O1, FoxO3, and FoxO4, and thereby modulates their activity. FoxOs restrain osteoclastogenesis and bone resorption. Here, we tested the hypothesis that the antiresorptive effects of Sirt1 are mediated by FoxOs. We report that Sirt1 activation by SRT2104 and SRT3025 inhibited murine osteoclast progenitor proliferation and reduced osteoclastogenesis. The effect of Sirt1 stimulators on osteoclastogenesis was abrogated in cells lacking FoxO1, FoxO3, and FoxO4. FoxO1 acetylation was increased by knocking down Sirt1 or addition of receptor activator of nuclear factor kappa-B ligand, the critical cytokine for osteoclast differentiation. Furthermore, acetylation inhibited, whereas deacetylation promoted, FoxO-mediated transcription. SRT3025 increased the expression of the FoxO-target genes catalase and hemeoxygenase-1 (HO-1) in osteoclast progenitors, in a FoxO-dependent manner. HO-1 catabolizes heme and attenuates mitochondrial oxidative phosphorylation and ATP production in macrophages. HO-1 levels were strongly reduced and ATP levels increased by Receptor activator of nuclear factor kappa-B ligand. In contrast, SRT3025 and FoxOs decreased ATP production, and the effect of SRT3025 was mediated by FoxOs. These findings reveal that the antiosteoclastogenic actions of Sirt1 are mediated by FoxOs and result from impaired mitochondria activity. Along with earlier findings that the osteoblastogenic effects of Sirt1 are also mediated by FoxOs, these results establish that the dual antiosteoporotic efficacy of Sirt1 stimulators (ie, decreasing bone resorption and promoting bone formation) is mediated via FoxO deacetylation.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Osteoclastos/metabolismo , Osteogênese , Sirtuína 1/metabolismo , Acetilação/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem da Célula/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ligante RANK/farmacologia , Células RAW 264.7 , Transcrição Gênica/efeitos dos fármacos
18.
J Bone Miner Res ; 30(7): 1138-49, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25704845

RESUMO

In men, androgens are critical for the acquisition and maintenance of bone mass in both the cortical and cancellous bone compartment. Male mice with targeted deletion of the androgen receptor (AR) in mature osteoblasts or osteocytes have lower cancellous bone mass, but no cortical bone phenotype. We have investigated the possibility that the effects of androgens on the cortical compartment result from AR signaling in osteoprogenitors or cells of the osteoclast lineage; or via estrogen receptor alpha (ERα) signaling in either or both of these two cell types upon conversion of testosterone to estradiol. To this end, we generated mice with targeted deletion of an AR or an ERα allele in the mesenchymal (AR(f/y);Prx1-Cre or ERα(f/f);Osx1-Cre) or myeloid cell lineage (AR(f/y);LysM-Cre or ERα(f/f);LysM-Cre) and their descendants. Male AR(f/y);Prx1-Cre mice exhibited decreased bone volume and trabecular number, and increased osteoclast number in the cancellous compartment. Moreover, they did not undergo the loss of cancellous bone volume and trabecular number caused by orchidectomy (ORX) in their littermate controls. In contrast, AR(f/y);LysM-Cre, ERα(f/f);Osx1-Cre, or ERα(f/f);LysM-Cre mice had no cancellous bone phenotype at baseline and lost the same amount of cancellous bone as their controls following ORX. Most unexpectedly, adult males of all four models had no discernible cortical bone phenotype at baseline, and lost the same amount of cortical bone as their littermate controls after ORX. Recapitulation of the effects of ORX by AR deletion only in the AR(f/y);Prx1-Cre mice indicates that the effects of androgens on cancellous bone result from AR signaling in osteoblasts-not on osteoclasts or via aromatization. The effects of androgens on cortical bone mass, on the other hand, do not require AR or ERα signaling in any cell type across the osteoblast or osteoclast differentiation lineage. Therefore, androgens must exert their effects indirectly by actions on some other cell type(s) or tissue(s).


Assuntos
Androgênios/farmacologia , Osso e Ossos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Osso e Ossos/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Deleção de Genes , Integrases/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Fenótipo , Microtomografia por Raio-X
19.
Nat Commun ; 5: 3773, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24781012

RESUMO

Besides their cell-damaging effects in the setting of oxidative stress, reactive oxygen species (ROS) play an important role in physiological intracellular signalling by triggering proliferation and survival. FoxO transcription factors counteract ROS generation by upregulating antioxidant enzymes. Here we show that intracellular H2O2 accumulation is a critical and purposeful adaptation for the differentiation and survival of osteoclasts, the bone cells responsible for the resorption of mineralized bone matrix. Using mice with conditional loss or gain of FoxO transcription factor function, or mitochondria-targeted catalase in osteoclasts, we demonstrate this is achieved, at least in part, by downregulating the H2O2-inactivating enzyme catalase. Catalase downregulation results from the repression of the transcriptional activity of FoxO1, 3 and 4 by RANKL, the indispensable signal for the generation of osteoclasts, via an Akt-mediated mechanism. Notably, mitochondria-targeted catalase prevented the loss of bone caused by loss of oestrogens, suggesting that decreasing H2O2 production in mitochondria may represent a rational pharmacotherapeutic approach to diseases with increased bone resorption.


Assuntos
Reabsorção Óssea/fisiopatologia , Catalase/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Peróxido de Hidrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Análise de Variância , Animais , Western Blotting , Células Cultivadas , Primers do DNA/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Regulação Enzimológica da Expressão Gênica/genética , Camundongos , Ligante RANK/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Microtomografia por Raio-X
20.
J Clin Invest ; 123(1): 394-404, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23221342

RESUMO

The detection of estrogen receptor-α (ERα) in osteoblasts and osteoclasts over 20 years ago suggested that direct effects of estrogens on both of these cell types are responsible for their beneficial effects on the skeleton, but the role of ERα in osteoblast lineage cells has remained elusive. In addition, estrogen activation of ERα in osteoclasts can only account for the protective effect of estrogens on the cancellous, but not the cortical, bone compartment that represents 80% of the entire skeleton. Here, we deleted ERα at different stages of differentiation in murine osteoblast lineage cells. We found that ERα in osteoblast progenitors expressing Osterix1 (Osx1) potentiates Wnt/ß-catenin signaling, thereby increasing proliferation and differentiation of periosteal cells. Further, this signaling pathway was required for optimal cortical bone accrual at the periosteum in mice. Notably, this function did not require estrogens. The osteoblast progenitor ERα mediated a protective effect of estrogens against endocortical, but not cancellous, bone resorption. ERα in mature osteoblasts or osteocytes did not influence cancellous or cortical bone mass. Hence, the ERα in both osteoblast progenitors and osteoclasts functions to optimize bone mass but at distinct bone compartments and in response to different cues.


Assuntos
Diferenciação Celular/fisiologia , Receptor alfa de Estrogênio/metabolismo , Osteoblastos/metabolismo , Periósteo/metabolismo , Células-Tronco/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Proliferação de Células , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Camundongos , Camundongos Mutantes , Osteoblastos/citologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Periósteo/citologia , Fator de Transcrição Sp7 , Células-Tronco/citologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA