Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Ophthalmol (Lausanne) ; 4: 1392106, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38984131

RESUMO

Aim: To evaluate the prevalence and clinical characteristics of limbal stem cell deficiency (LSCD) in the setting of a tertiary referral cornea practice at an academic center. Patient and methods: A retrospective chart review was performed to identify all unique medical record numbers (MRNs) presenting to a single cornea specialist (JHH) at the University of Minnesota during calendar years 2019 and 2020. Records were queried and confirmed for a diagnosis of LSCD. Clinical characteristics of identified patients, including demographics, etiology of LSCD, severity of LSCD, treatment, and best corrected visual acuity (BCVA) at final follow-up, were documented. Results: In total 1436 unique MRNs were identified over the study period. There were 61 individuals (91 eyes) diagnosed with LSCD, resulting in a prevalence of 4.25% (95% CI, 3.33-5.42). Of 91 eyes, 60 eyes were bilateral (65.9%). Among all eyes, ocular surface burns were the most common etiology (18.7%) followed by iatrogenic or medicamentosa (15.4%). There were 51 eyes (56.0%) that underwent some form of transplantation. The median BCVA at final follow-up was Snellen 20/80 (range 20/20 to no light perception). Conclusions: The prevalence of LSCD found at a cornea subspecialty tertiary referral center in our study was much higher than previously reported prevalence rates. This may reflect referral bias and potential underdiagnosis of LSCD in practices outside of subspecialty referral centers. The high prevalence rate in our study also suggests that LSCD patients are concentrated in subspecialty referral practices, with many having high morbidity disease. This constitutes a major health burden for these practices.

2.
J Periodontol ; 92(10): 1483-1495, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33341947

RESUMO

BACKGROUND: Coinciding with other chronic comorbidities, the prevalence of periodontal disease increases with aging. Mounting evidence has established that senescent cells accumulate at sites of age-related pathologies, where they promote "non-microbial" inflammation. We hypothesized that alveolar bone osteocytes develop senescence characteristics in old age. METHODS: Alveolar bone samples were obtained from young (6 months) and old (20 to 22 months) mice to evaluate the expression of senescence biomarkers by immunofluorescent staining. Osteocyte-enriched fractions were used to characterize the age-related senescence-associated secretory phenotype (SASP) gene expression profile. Primary alveolar bone cells were exposed to the SASP via in vitro senescent conditioned media (SCM) administration. A multiplex assay confirmed protein levels of specific cytokines. Interactions with bacterial components were evaluated by stimulating cells with lipopolysaccharide (LPS). RESULTS: Increased senescence-associated distension of satellites (SADS) and p16Ink4a mRNA expression were identified in alveolar bone osteocytes with aging. These findings were associated with increased levels of DNA damage, and activated p38 MAPK, both inducers of senescence. Furthermore, interleukin-6 (IL6), IL17, IGFBP4, and MMP13 were significantly upregulated with aging in osteocyte-enriched samples. Interestingly, SCM potentiated the LPS-induced expression of IL1α, IL1ß, and IL6. Cell migration and differentiation were also impeded by SCM. These in vitro effects were ameliorated by the p38 MAPK inhibitor SB202190. CONCLUSIONS: Accumulation of senescent osteocytes contributes to deterioration of the periodontal environment by exacerbating chronic inflammation and reducing regeneration in old age. Cellular senescence is a cell-intrinsic response to DNA damage, and a host-related mechanism associated with aging that could potentiate inflammation induced by bacterial components.


Assuntos
Senescência Celular , Doenças Periodontais , Envelhecimento , Animais , Progressão da Doença , Inflamação , Camundongos , Osteócitos
3.
JCI Insight ; 5(9)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32267250

RESUMO

The worldwide prevalence of type 2 diabetes (T2D) is increasing. Despite normal to higher bone density, patients with T2D paradoxically have elevated fracture risk resulting, in part, from poor bone quality. Advanced glycation endproducts (AGEs) and inflammation as a consequence of enhanced receptor for AGE (RAGE) signaling are hypothesized culprits, although the exact mechanisms underlying skeletal dysfunction in T2D are unclear. Lack of inducible models that permit environmental (in obesity) and temporal (after skeletal maturity) control of T2D onset has hampered progress. Here, we show in C57BL/6 mice that a onetime pharmacological intervention (streptozotocin, STZ) initiated in adulthood combined with high-fat diet-induced (HFD-induced) obesity caused hallmark features of human adult-onset T2D, including prolonged hyperglycemia, insulin resistance, and pancreatic ß cell dysfunction, but not complete destruction. In addition, HFD/STZ (i.e., T2D) resulted in several changes in bone quality that closely mirror those observed in humans, including compromised bone microarchitecture, reduced biomechanical strength, impaired bone material properties, altered bone turnover, and elevated levels of the AGE CML in bone and blood. Furthermore, T2D led to the premature accumulation of senescent osteocytes with a unique proinflammatory signature. These findings highlight the RAGE pathway and senescent cells as potential targets to treat diabetic skeletal fragility.


Assuntos
Osso e Ossos , Diabetes Mellitus Tipo 2/metabolismo , Osteócitos , Animais , Densidade Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Senescência Celular , Modelos Animais de Doenças , Produtos Finais de Glicação Avançada/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/metabolismo , Osteócitos/patologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo
4.
Bone ; 132: 115220, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31904537

RESUMO

Cellular senescence is associated with inflammation and extracellular matrix tissue remodeling through the secretion of proteins termed the senescence-associated secretory phenotype (SASP). Although osteocyte senescence in older individuals in the skeleton is well recognized, whether young alveolar osteocytes can also become senescent is unknown. This is potentially important in the context of periodontal disease, which is an inflammatory condition caused by a gradual change from symbiotic to pathogenic oral microflora that can lead to tooth loss. Our aim was to identify whether senescent osteocytes accumulate in young alveolar bone and whether bacterial-derived lipopolysaccharide (LPS) can influence cellular senescence in alveolar bone. An osteocyte-enriched cell population isolated from alveolar bone expressed increased levels of the known senescence marker p16Ink4a, as well as select SASP markers known to be implicated alveolar bone resorption (Icam1, Il6, Il17, Mmp13 and Tnfα), compared to ramus control cells. Increased senescence of alveolar bone osteocytes was also observed in vivo using the senescence-associated distension of satellites (SADS) assay and increased γH2AX, a marker of DNA damage associated with senescent cells. To approximate a bacterial infection in vitro, alveolar osteocytes were treated with LPS. We found increased expression of various senescence and SASP markers, increased γH2AX staining, increased SA-ß-Gal activity and the redistribution of F-actin leading to a larger and flattened cell morphology, all hallmarks of cellular senescence. In conclusion, our data suggests a model whereby bacterial-derived LPS stimulates premature alveolar osteocyte senescence, which in combination with the resultant SASP, could potentially contribute to the onset of alveolar bone loss.


Assuntos
Perda do Osso Alveolar , Osteócitos , Idoso , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina , Humanos , Lipopolissacarídeos/toxicidade
5.
J Bone Miner Res ; 34(8): 1407-1418, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30913313

RESUMO

Estrogen deficiency is a seminal mechanism in the pathogenesis of osteoporosis. Mounting evidence, however, establishes that cellular senescence, a fundamental mechanism that drives multiple age-related diseases, also causes osteoporosis. Recently, we systematically identified an accumulation of senescent cells, characterized by increased p16Ink4a and p21Cip1 levels and development of a senescence-associated secretory phenotype (SASP), in mouse bone/marrow and human bone with aging. We then demonstrated that elimination of senescent cells prevented age-related bone loss using multiple approaches, eg, treating old mice expressing a "suicide" transgene, INK-ATTAC, with AP20187 to induce apoptosis of p16Ink4a -senescent cells or periodically treating old wild-type mice with "senolytics," ie, drugs that eliminate senescent cells. Here, we investigate a possible role for estrogen in the regulation of cellular senescence using multiple approaches. First, sex steroid deficiency 2 months after ovariectomy (OVX, n = 15) or orchidectomy (ORCH, n = 15) versus sham surgery (SHAM, n = 15/sex) in young adult (4-month-old) wild-type mice did not alter senescence biomarkers or induce a SASP in bone. Next, in elderly postmenopausal women, 3 weeks of estrogen therapy (n = 10; 74 ± 5 years) compared with no treatment (n = 10; 78 ± 5 years) did not alter senescence biomarkers or the SASP in human bone biopsies. Finally, young adult (4-month-old) female INK-ATTAC mice were randomized (n = 17/group) to SHAM+Vehicle, OVX+Vehicle, or OVX+AP20187 for 2 months. As anticipated, OVX+Vehicle caused significant trabecular/cortical bone loss compared with SHAM+Vehicle. However, treatment with AP20187, which eliminates senescent cells in INK-ATTAC mice, did not rescue the OVX-induced bone loss or alter senescence biomarkers. Collectively, our data establish independent roles of estrogen deficiency and cellular senescence in the pathogenesis of osteoporosis, which has important implications for testing novel senolytics for skeletal efficacy, as these drugs will need to be evaluated in preclinical models of aging as opposed to the current FDA model of prevention of OVX-induced bone loss. © 2019 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento , Osso Esponjoso , Senescência Celular , Estrogênios/deficiência , Osteoporose , Adulto , Idoso , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Osso Esponjoso/metabolismo , Osso Esponjoso/patologia , Estrogênios/metabolismo , Feminino , Humanos , Masculino , Camundongos , Osteoporose/metabolismo , Osteoporose/patologia , Ovariectomia
6.
J Bone Miner Res ; 34(1): 135-144, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30321475

RESUMO

Developing novel approaches to treat skeletal disorders requires an understanding of how critical molecular factors regulate osteoblast differentiation and bone remodeling. We have reported that (1) retinoic acid receptor-related orphan receptor beta (Rorß) is upregulated in bone samples isolated from aged mice and humans in vivo; (2) Rorß expression is inhibited during osteoblastic differentiation in vitro; and (3) genetic deletion of Rorß in mice results in preservation of bone mass during aging. These data establish that Rorß inhibits osteogenesis and that strict control of Rorß expression is essential for bone homeostasis. Because microRNAs (miRNAs) are known to play important roles in the regulation of gene expression in bone, we explored whether a predicted subset of nine miRNAs regulates Rorß expression during both osteoblast differentiation and aging. Mouse osteoblastic cells were differentiated in vitro and assayed for Rorß and miRNA expression. As Rorß levels declined with differentiation, the expression of many of these miRNAs, including miR-219a-5p, was increased. We further demonstrated that miR-219a-5p was decreased in bone samples from old (24-month) mice, as compared with young (6-month) mice, concomitant with increased Rorß expression. Importantly, we also found that miR-219a-5p expression was decreased in aged human bone biopsies compared with young controls, demonstrating that this phenomenon also occurs in aging bone in humans. Inhibition of miR-219a-5p in mouse calvarial osteoblasts led to increased Rorß expression and decreased alkaline phosphatase expression and activity, whereas a miR-219a-5p mimic decreased Rorß expression and increased osteogenic activity. Finally, we demonstrated that miR-219a-5p physically interacts with Rorß mRNA in osteoblasts, defining Rorß as a true molecular target of miR-219a-5p. Overall, our findings demonstrate that miR-219a-5p is involved in the regulation of Rorß in both mouse and human bone. © 2018 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento , Diferenciação Celular , Regulação da Expressão Gênica , MicroRNAs/metabolismo , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Osteoblastos/metabolismo , Osteoporose/metabolismo , Animais , Humanos , Camundongos , MicroRNAs/genética , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Osteoblastos/patologia , Osteoporose/genética , Osteoporose/patologia
7.
Nat Med ; 24(8): 1246-1256, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29988130

RESUMO

Physical function declines in old age, portending disability, increased health expenditures, and mortality. Cellular senescence, leading to tissue dysfunction, may contribute to these consequences of aging, but whether senescence can directly drive age-related pathology and be therapeutically targeted is still unclear. Here we demonstrate that transplanting relatively small numbers of senescent cells into young mice is sufficient to cause persistent physical dysfunction, as well as to spread cellular senescence to host tissues. Transplanting even fewer senescent cells had the same effect in older recipients and was accompanied by reduced survival, indicating the potency of senescent cells in shortening health- and lifespan. The senolytic cocktail, dasatinib plus quercetin, which causes selective elimination of senescent cells, decreased the number of naturally occurring senescent cells and their secretion of frailty-related proinflammatory cytokines in explants of human adipose tissue. Moreover, intermittent oral administration of senolytics to both senescent cell-transplanted young mice and naturally aged mice alleviated physical dysfunction and increased post-treatment survival by 36% while reducing mortality hazard to 65%. Our study provides proof-of-concept evidence that senescent cells can cause physical dysfunction and decreased survival even in young mice, while senolytics can enhance remaining health- and lifespan in old mice.


Assuntos
Dasatinibe/farmacologia , Longevidade/efeitos dos fármacos , Quercetina/farmacologia , Tecido Adiposo/metabolismo , Animais , Transplante de Células , Senescência Celular/efeitos dos fármacos , Citocinas/metabolismo , Dieta Hiperlipídica , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Estresse Fisiológico/efeitos dos fármacos , Análise de Sobrevida
8.
J Bone Miner Res ; 33(4): 720-731, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29206307

RESUMO

There is a clinical need to identify new molecular targets for the treatment of osteoporosis, particularly those that simultaneously inhibit bone resorption while stimulating bone formation. We have previously shown in overexpression studies that retinoic acid receptor-related orphan receptor ß (Rorß) suppresses in vitro osteoblast differentiation. In addition, the expression of Rorß is markedly increased in bone marrow-derived mesenchymal stromal cells with aging in both mice and humans. Here we establish a critical role for Rorß in regulating bone metabolism using a combination of in vitro and in vivo studies. We used Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 gene editing to demonstrate that loss of Rorß in osteoblasts enhances Wnt signaling, specifically through increased recruitment of ß-catenin to T-cell factor/lymphoid enhancer factor (Tcf/Lef) DNA binding sites in the promoters of the Wnt target genes Tcf7 and Opg. This resulted in increased osteogenic gene expression and suppressed osteoclast formation through increased osteoprotegerin (OPG) secretion in Rorß-deficient cells. Consistent with our in vitro data, genetic deletion of Rorß in both female and male mice resulted in preserved bone mass and microarchitecture with advancing age due to increased bone formation with a concomitant decrease in resorption. The improved skeletal phenotype in the Rorß-/- mice was also associated with increased bone protein levels of TCF7 and OPG. These data demonstrate that loss of Rorß has beneficial skeletal effects by increasing bone formation and decreasing bone resorption, at least in part through ß-catenin-dependent activation of the Wnt pathway. Thus, inhibition of Rorß represents a novel approach to potentially prevent or reverse osteoporosis. © 2017 American Society for Bone and Mineral Research.


Assuntos
Reabsorção Óssea/metabolismo , Diferenciação Celular , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/deficiência , Osteoblastos/metabolismo , Osteogênese , Via de Sinalização Wnt , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Linhagem Celular , Camundongos , Camundongos Knockout , Osteoblastos/patologia
10.
Nat Med ; 23(9): 1072-1079, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28825716

RESUMO

Aging is associated with increased cellular senescence, which is hypothesized to drive the eventual development of multiple comorbidities. Here we investigate a role for senescent cells in age-related bone loss through multiple approaches. In particular, we used either genetic (i.e., the INK-ATTAC 'suicide' transgene encoding an inducible caspase 8 expressed specifically in senescent cells) or pharmacological (i.e., 'senolytic' compounds) means to eliminate senescent cells. We also inhibited the production of the proinflammatory secretome of senescent cells using a JAK inhibitor (JAKi). In aged (20- to 22-month-old) mice with established bone loss, activation of the INK-ATTAC caspase 8 in senescent cells or treatment with senolytics or the JAKi for 2-4 months resulted in higher bone mass and strength and better bone microarchitecture than in vehicle-treated mice. The beneficial effects of targeting senescent cells were due to lower bone resorption with either maintained (trabecular) or higher (cortical) bone formation as compared to vehicle-treated mice. In vitro studies demonstrated that senescent-cell conditioned medium impaired osteoblast mineralization and enhanced osteoclast-progenitor survival, leading to increased osteoclastogenesis. Collectively, these data establish a causal role for senescent cells in bone loss with aging, and demonstrate that targeting these cells has both anti-resorptive and anabolic effects on bone. Given that eliminating senescent cells and/or inhibiting their proinflammatory secretome also improves cardiovascular function, enhances insulin sensitivity, and reduces frailty, targeting this fundamental mechanism to prevent age-related bone loss suggests a novel treatment strategy not only for osteoporosis, but also for multiple age-related comorbidities.


Assuntos
Osso e Ossos/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Janus Quinases/antagonistas & inibidores , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Osteoporose/metabolismo , Pirazóis/farmacologia , Absorciometria de Fóton , Animais , Apoptose/genética , Osso e Ossos/metabolismo , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/metabolismo , Caspase 8/genética , Diferenciação Celular , Senescência Celular/genética , Osso Cortical/efeitos dos fármacos , Osso Cortical/metabolismo , Meios de Cultivo Condicionados , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Nitrilas , Osteoblastos/citologia , Osteoclastos/citologia , Osteoporose/genética , Pirimidinas , Reação em Cadeia da Polimerase em Tempo Real , Suporte de Carga , beta-Galactosidase
11.
J Bone Miner Res ; 31(11): 1920-1929, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27341653

RESUMO

Cellular senescence is a fundamental mechanism by which cells remain metabolically active yet cease dividing and undergo distinct phenotypic alterations, including upregulation of p16Ink4a , profound secretome changes, telomere shortening, and decondensation of pericentromeric satellite DNA. Because senescent cells accumulate in multiple tissues with aging, these cells and the dysfunctional factors they secrete, termed the senescence-associated secretory phenotype (SASP), are increasingly recognized as promising therapeutic targets to prevent age-related degenerative pathologies, including osteoporosis. However, the cell type(s) within the bone microenvironment that undergoes senescence with aging in vivo has remained poorly understood, largely because previous studies have focused on senescence in cultured cells. Thus in young (age 6 months) and old (age 24 months) mice, we measured senescence and SASP markers in vivo in highly enriched cell populations, all rapidly isolated from bone/marrow without in vitro culture. In both females and males, p16Ink4a expression by real-time quantitative polymerase chain reaction (rt-qPCR) was significantly higher with aging in B cells, T cells, myeloid cells, osteoblast progenitors, osteoblasts, and osteocytes. Further, in vivo quantification of senescence-associated distension of satellites (SADS), ie, large-scale unraveling of pericentromeric satellite DNA, revealed significantly more senescent osteocytes in old compared with young bone cortices (11% versus 2%, p < 0.001). In addition, primary osteocytes from old mice had sixfold more (p < 0.001) telomere dysfunction-induced foci (TIFs) than osteocytes from young mice. Corresponding with the age-associated accumulation of senescent osteocytes was significantly higher expression of multiple SASP markers in osteocytes from old versus young mice, several of which also showed dramatic age-associated upregulation in myeloid cells. These data show that with aging, a subset of cells of various lineages within the bone microenvironment become senescent, although senescent myeloid cells and senescent osteocytes predominantly develop the SASP. Given the critical roles of osteocytes in orchestrating bone remodeling, our findings suggest that senescent osteocytes and their SASP may contribute to age-related bone loss. © 2016 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/citologia , Microambiente Celular , Senescência Celular , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA Satélite/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteócitos/metabolismo , Fenótipo
12.
J Bone Miner Res ; 31(3): 606-14, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26418452

RESUMO

Although the role of ERα in regulating bone metabolism has been extensively studied, ERß has been largely dismissed as a relevant modulator of bone mass. Previous studies examining ERß utilized a germline knockout mouse expressing transcript variants of ERß and displaying systemic hormonal changes that confounded interpretation of the skeletal phenotype. Thus, we used a conditional ERß mouse model to achieve deletion of ERß specifically in early osteoprogenitor cells using the Prx1-Cre driver. We observed marked increases in the trabecular bone volume fraction (of 58% [p < 0.003] and 93% [p < 0.0003] in 6- and 12-week-old female ERß(Prx1-CKO) mice, respectively) but no changes in cortical bone. Serum estradiol and IGF-I levels were unaltered in ERß(Prx1-CKO) mice. Bone formation and resorption indices by histomorphometry and serum assays were unchanged in these mice, suggesting that alterations in bone turnover may have occurred early in development. However, the ratio of colony-forming unit-osteoblasts (CFU-OBs) to CFU-fibroblasts (CFU-Fs) was increased in bone marrow cultures from ERß(Prx1-CKO) compared with control mice, indicating increased differentiation of osteoblast precursor cells into osteoblasts in ERß(Prx1-CKO) mice. Detailed quantitative polymerase chain reaction analyses of 128 genes in 16 prespecified pathways revealed significant downregulation of 11 pathways in ERß(Prx1-CKO) mice. Thus, deletion of ERß specifically in osteoblast lineage cells, in the absence of all splice variants, increases trabecular bone mass and modulates multiple pathways related to bone metabolism. These findings suggest that pharmacological inhibition of ERß in bone may provide a novel approach to treat osteoporosis.


Assuntos
Osso e Ossos/citologia , Osso Esponjoso/anatomia & histologia , Osso Cortical/anatomia & histologia , Receptor beta de Estrogênio/metabolismo , Deleção de Genes , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Osteoblastos/metabolismo , Ovariectomia , Microtomografia por Raio-X
13.
J Cell Biochem ; 116(1): 58-66, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25145595

RESUMO

Hematopoietic stem cell (HSC) self-renewal is regulated by osteoblast and/or endothelial cells within the hematopoietic niche. However, the true identity of the supporting cells and the nature of the secreted factors remain uncertain. We developed a novel mouse model and analyzed whether circulating human peripheral hematopoietic lineage negative/AP+ (lin-/AP+) cells support hematopoiesis in vivo. Thus, immunocompromised (Rag) mice expressing thymidine kinase (Tk) under the control of the 3.6Col1α1 promoter (Tk-Rag) were treated with ganciclovir, resulting in osteoblast progenitor cell ablation and subsequent loss of hematopoiesis (evaluated by measuring mouse Ter119+ erythroid cells). Following hematopoietic cell depletion, human bone marrow-derived marrow stromal cells (MSCs) or lin-/AP+ cells were infused into Tk-Rag mice and compared with saline infusions. Ganciclovir significantly reduced (7.4-fold) Ter119+ cells in the bone marrow of Tk-Rag mice compared to saline injections. Infusion of either MSCs or lin-/AP+ cells into ganciclovir-treated mice resulted in a 3.3-fold and 2.7-fold increase (P < 0.01), respectively, in Ter119+ cells compared to mice receiving saline. Relative to lin-/AP- cells, lin-/AP+ cells expressed high levels of mesenchymal, endothelial, and hematopoiesis supporting genes. Thus, human peripheral blood lin-/AP+ cells represent a novel cell type capable of supporting hematopoiesis in a manner comparable to MSCs.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Linhagem da Célula , Feminino , Citometria de Fluxo , Ganciclovir/farmacologia , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos
14.
Analyst ; 138(16): 4470-4, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23775114

RESUMO

Prior to exhibiting an African Komo mask from the collections of the Detroit Institute of Arts, a multianalytical approach was undertaken to characterize the flaking encrusted coating on the surface of the mask. Preliminary XRF and FTIR examination of the coating on the Komo mask revealed the presence of significant quantities of iron and protein, possibly indicating the presence of blood. Raman spectroscopy showed evidence for the porphyrin structure of haem as well. To confirm that blood was indeed present in the coating, we developed a novel method for identifying the haem moiety from blood by use of in situ methylation and direct analysis in real time mass spectrometry (DART-MS). Following a denaturing step with formic acid, the resulting solution was combined with an excess of phenyltrimethylammonium hydroxide to promote desorption, applied to a melting point tube, and placed into the direct analysis in real time ion source gas stream at 550 °C. The permethylated haem ion (m/z 644.208) from myoglobin, haemoglobin, fresh blood, and blood aged in the laboratory for 10 years was readily observed above the background. By the described DART-TOF-MS method, permethylated haem was positively identified in the mask coating, confirming the presence of blood. This method has obvious utility in forensic science beyond that for identifying blood incorporated in cultural heritage materials.


Assuntos
Comportamento Ritualístico , Sistemas Computacionais , Teste em Amostras de Sangue Seco/métodos , Heme/análise , Espectrometria de Massas/métodos , Museus , África , Animais , Bovinos , Heme/química , Humanos , Mali , Suínos
15.
Endocrinology ; 153(11): 5325-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23015293

RESUMO

The mechanisms of estrogen receptor (ER)-α activity can be categorized into those involving direct (classical) or indirect (nonclassical) DNA binding. Although various mouse models have demonstrated the importance of ERα in bone, the specific gene expression patterns affected by these modes of ERα action are unknown. In this report, the gene expression patterns of ERα-deficient (ERKO) mice and nonclassical ER knock-in (NERKI) mice, which can function only by nonclassical means, were analyzed. Three-month-old mice were ovariectomized and implanted with estrogen pellets for 1 month to normalize estrogen levels. Microarray analysis of flushed cortical bone revealed 28% (210 of 763) of the genes differentially expressed in ERKO mice were altered in NERKI mice, suggesting estrogen response element-dependent regulation of these genes in bone. Pathway analysis revealed alterations in genes involved in focal adhesion and extracellular matrix interactions. However, the majority of genes regulated in ERKO mice (72%) were unique (i.e. not altered in NERKI mice), suggesting these are regulated by nonclassical mechanisms. To further explore the pathways affected in ERKO mice, we performed focused quantitative PCR arrays for genes involved in various aspects of bone physiology. Genes involved in bone formation, senescence, apoptosis, and autophagy were significantly regulated. Overall, the majority of the genes regulated by ERα in bone are via nonclassical pathways. However, because NERKI mice display an osteoporotic phenotype, it can be deduced that the minority of the estrogen response element-dependent genes/pathways play critical roles in the regulation of bone physiology. These data demonstrate the importance of classical ERα signaling in regulating bone metabolism.


Assuntos
Osso e Ossos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Osso e Ossos/efeitos dos fármacos , Estradiol/sangue , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Estrogênios/sangue , Estrogênios/farmacologia , Feminino , Camundongos , Camundongos Transgênicos , Ovariectomia , Transdução de Sinais/efeitos dos fármacos
16.
J Orthop Res ; 29(12): 1804-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21674605

RESUMO

Development of novel therapeutic approaches to repair fracture non-unions remains a critical clinical necessity. We evaluated the capacity of human embryonic stem cell (hESC)-derived mesenchymal stem/stromal cells (MSCs) to induce healing in a fracture non-union model in rats. In addition, we placed these findings in the context of parallel studies using human bone marrow MSCs (hBM-MSCs) or a no cell control group (n = 10-12 per group). Preliminary studies demonstrated that both for hESC-derived MSCs and hBM-MSCs, optimal induction of fracture healing required in vitro osteogenic differentiation of these cells. Based on biomechanical testing of fractured femurs, maximum torque, and stiffness were significantly greater in the hBM-MSC as compared to the control group that received no cells; values for these parameters in the hESC-derived MSC group were intermediate between the hBM-MSC and control groups, and not significantly different from the control group. However, some evidence of fracture healing was evident by X-ray in the hESC-derived MSC group. Our results thus indicate that while hESC-derived MSCs may have potential to induce fracture healing in non-unions, hBM-MSCs function more efficiently in this process. Additional studies are needed to further modify hESCs to achieve optimal fracture healing by these cells.


Assuntos
Transplante de Medula Óssea/métodos , Fraturas do Fêmur/terapia , Consolidação da Fratura/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Células Estromais/transplante , Animais , Células da Medula Óssea/citologia , Calo Ósseo/diagnóstico por imagem , Linhagem Celular , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Fraturas do Fêmur/diagnóstico por imagem , Fibroblastos/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Radiografia , Ratos , Ratos Nus , Células Estromais/citologia , Transplante Heterólogo
17.
Bone ; 49(2): 208-16, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21458604

RESUMO

Estrogen receptor (ER) α is a major regulator of bone metabolism which can modulate gene expression via a "classical" pathway involving direct DNA binding to estrogen-response elements (EREs) or via "non-classical" pathways involving protein-protein interactions. While the skeletal consequences of loss of ERE binding by ERα have been described, a significant unresolved question is how loss of ERE binding differs from complete loss of ERα. Thus, we compared the skeletal phenotype of wild-type (ERα(+/+)) and ERα knock out (ERα(-/-)) mice with that of mice in which the only ERα present had a knock-in mutation abolishing ERE binding (non-classical ERα knock-in [NERKI], ERα(-/NERKI)). All three groups were in the same genetic background (C57BL/6). As compared to both ERα(+/+) and ERα(-/-) mice, ERα(-/NERKI) mice had significantly reduced cortical volumetric bone mineral density and thickness at the tibial diaphysis; this was accompanied by significant decreases in periosteal and endocortical mineral apposition rates. Colony forming unit (CFU)-fibroblast, CFU-alkaline phosphatase, and CFU-osteoblast numbers were all increased in ERα(-/-) compared to ERα(+/+) mice, but reduced in ERα(-/NERKI) mice compared to the two other groups. Thus, using mice in identical genetic backgrounds, our data indicate that the presence of an ERα that cannot bind DNA but can function through protein-protein interactions may have more deleterious skeletal effects than complete loss of ERα. These findings suggest that shifting the balance of classical versus non-classical ERα signaling triggers pathways that impair bone formation. Further studies defining these pathways may lead to novel approaches to selectively modulate ER signaling for beneficial skeletal effects.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Receptor alfa de Estrogênio/deficiência , Receptores de Estrogênio/metabolismo , Transdução de Sinais/fisiologia , Absorciometria de Fóton , Adipócitos/citologia , Animais , Composição Corporal/genética , Composição Corporal/fisiologia , Peso Corporal/genética , Peso Corporal/fisiologia , Densidade Óssea/genética , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Estradiol/sangue , Receptor alfa de Estrogênio/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Osteoblastos/citologia , Radioimunoensaio , Receptores de Estrogênio/genética , Transdução de Sinais/genética , Microtomografia por Raio-X
18.
J Bone Miner Res ; 25(11): 2438-46, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20499336

RESUMO

While female mice do not have the equivalent of a menopause, they do undergo reproductive senescence. Thus, to dissociate the effects of aging versus estrogen deficiency on age-related bone loss, we sham-operated, ovariectomized, or ovariectomized and estrogen-replaced female C57/BL6 mice at 6 months of age and followed them to age 18 to 22 months. Lumbar spines and femurs were excised for analysis, and bone marrow hematopoietic lineage negative (lin-) cells (enriched for osteoprogenitor cells) were isolated for gene expression studies. Six-month-old intact control mice were euthanized to define baseline parameters. Compared with young mice, aged/sham-operated mice had a 42% reduction in lumbar spine bone volume/total volume (BV/TV), and maintaining constant estrogen levels over life in ovariectomized/estrogen-treated mice did not prevent age-related trabecular bone loss at this site. By contrast, lifelong estrogen treatment of ovariectomized mice completely prevented the age-related reduction in cortical volumetric bone mineral density (vBMD) and thickness at the tibial diaphysis present in the aged/sham-operated mice. As compared with cells from young mice, lin- cells from aged/sham-operated mice expressed significantly higher mRNA levels for osteoblast differentiation and proliferation marker genes. These data thus demonstrate that, in mice, age-related loss of cortical bone in the appendicular skeleton, but not loss of trabecular bone in the spine, can be prevented by maintaining constant estrogen levels over life. The observed increase in osteoblastic differentiation and proliferation marker gene expression in progenitor bone marrow cells from aged versus young mice may represent a compensatory mechanism in response to ongoing bone loss.


Assuntos
Estrogênios/uso terapêutico , Osteoporose/tratamento farmacológico , Absorciometria de Fóton , Animais , Peso Corporal/efeitos dos fármacos , Contagem de Células , Estrogênios/farmacologia , Feminino , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoporose/diagnóstico por imagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Tomografia Computadorizada por Raios X
19.
J Biol Chem ; 284(28): 18767-77, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19423703

RESUMO

Both estrogen receptor (ER) and peroxisome proliferator-activated receptor gamma (PPARgamma) regulate bone metabolism, and because steroid receptor coactivator (SRC)-2 (TIF-2) enhances ER and PPARgamma activity, we examined the consequences of deletion of SRC-2 on bone using SRC-2 knock out (KO) mice. Loss of SRC-2 resulted in increased bone mass, with SRC-2 KO mice having 80% higher trabecular bone volume as compared with wild type mice. SRC-2 KO mice also had a marked decrease (by 50%) in bone marrow adipocytes. These data suggested that marrow precursor cells in the SRC-2 KO mice may be resistant to the inhibitory effects of endogenous PPARgamma ligands on bone formation. Consistent with this, compared with cultures from wild type mice, marrow stromal cultures from SRC-2 KO mice formed significantly more mineralized nodules (by 3-fold) in the presence of the PPARgamma agonist, rosiglitazone. Using chromatin immunoprecipitation analysis, we demonstrated that in bone marrow stromal cells, loss of SRC-2 leads to destabilization of the transcription complex at the peroxisome proliferator response elements of a number of PPARgamma target genes, resulting in an overall decrease in the expression of adipocyte-related genes and a marked decrease in adipocyte development. Using ovariectomy with or without estrogen replacement, we also demonstrated that SRC-2 KO mice were partially resistant to the skeletal actions of estrogen. Collectively, these findings indicate that loss of SRC-2 leads to partial skeletal resistance to the ER and PPARgamma, but resistance to PPARgamma is dominant, leading to increased bone mass. Modulating SRC-2 action may, thus, represent a novel therapeutic target for osteoporosis.


Assuntos
Deleção de Genes , Regulação da Expressão Gênica , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/fisiologia , PPAR gama/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Células da Medula Óssea/citologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Células Cultivadas , Densitometria , Feminino , Camundongos , Camundongos Knockout , Modelos Biológicos , Osteoporose/terapia , Rosiglitazona , Tiazolidinedionas/farmacologia , Tomografia Computadorizada por Raios X/métodos
20.
Endocrinology ; 148(4): 1902-10, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17204553

RESUMO

The role of estrogen signaling in the male skeleton via estrogen receptor (ER)-alpha is now well established. ERalpha can elicit responses through either classical estrogen response elements (ERE) pathways or nonclassical, non-ERE pathways. In the present study, we examined the effects of either the attenuation or loss of classical ERalpha signaling on the murine male skeleton. To accomplish this, we crossed male mice heterozygous for a knock-in mutation [nonclassical ERalpha knock-in (NERKI)], which abolishes the ERE-mediated pathway with female heterozygous ERalpha knockout mice (ERalpha+/-) and studied the F1 generation ERalpha+/+, ERalpha+/-, ERalpha+/NERKI, and ERalpha-/NERKI male progeny longitudinally using bone density and histomorphometry. The only ERalpha allele present in ERalpha-/NERKI mice is incapable of classical ERE-mediated signaling, whereas the heterozygous ERalpha+/NERKI mice have both one intact ERalpha and one NERKI allele. As compared with ERalpha+/+ littermates (n=10/genotype), male ERalpha+/NERKI and ERalpha-/NERKI mice displayed axial and appendicular skeletal osteopenia at 6, 12, 20, and 25 wk of age, as demonstrated by significant reductions in total bone mineral density (BMD) at representative sites (areal BMD by dual-energy x-ray absorptiometry at the lumbar vertebrae and femur and volumetric BMD by peripheral quantitative computed tomography at the tibia; P<0.05-0.001 vs. ERalpha+/+). The observed osteopenia in these mice was evident in both trabecular and cortical bone compartments. However, these decreases were more severe in mice lacking classical ERalpha signaling (ERalpha-/NERKI mice), compared with mice in which one wild-type ERalpha allele was present (ERalpha+/NERKI mice). Collectively, these data demonstrate that classical ERalpha signaling is crucial for the development of the murine male skeleton.


Assuntos
Osso e Ossos/anatomia & histologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Elementos de Resposta/fisiologia , Animais , Peso Corporal , Densidade Óssea , Desenvolvimento Ósseo/genética , Osso e Ossos/metabolismo , Força Compressiva , Feminino , Vértebras Lombares/anatomia & histologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...