Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37560905

RESUMO

Background: The clinical healing environment after a posterior spinal arthrodesis surgery is one of the most clinically challenging bone-healing environments across all orthopedic interventions due to the absence of a contained space and the need to form de novo bone. Our group has previously reported that sclerostin in expressed locally at high levels throughout a developing spinal fusion. However, the role of sclerostin in controlling bone fusion remains to be established. Methods: We computationally identified two FDA-approved drugs, as well as a single novel small-molecule drug, for their ability to disrupt the interaction between sclerostin and its receptor, LRP5/6. The drugs were tested in several in vitro biochemical assays using murine MC3T3 and MSCs, assessing their ability to (1) enhance canonical Wnt signaling, (2) promote the accumulation of the active (non-phosphorylated) form of ß-catenin, and (3) enhance the intensity and signaling duration of BMP signaling. These drugs were then tested subcutaneously in rats as standalone osteoinductive agents on plain collagen sponges. Finally, the top drug candidates (called VA1 and C07) were tested in a rabbit posterolateral spine fusion model for their ability to achieve a successful fusion at 6 wk. Results: We show that by controlling GSK3b phosphorylation our three small-molecule inhibitors (SMIs) simultaneously enhance canonical Wnt signaling and potentiate canonical BMP signaling intensity and duration. We also demonstrate that the SMIs produce dose-dependent ectopic mineralization in vivo in rats as well as significantly increase posterolateral spine fusion rates in rabbits in vivo, both as standalone osteogenic drugs and in combination with autologous iliac crest bone graft. Conclusions: Few if any osteogenic small molecules possess the osteoinductive potency of BMP itself - that is, the ability to form de novo ectopic bone as a standalone agent. Herein, we describe two such SMIs that have this unique ability and were shown to induce de novo bone in a stringent in vivo environment. These SMIs may have the potential to be used in novel, cost-effective bone graft substitutes for either achieving spinal fusion or in the healing of critical-sized fracture defects. Funding: This work was supported by a Veteran Affairs Career Development Award (IK2-BX003845).


Assuntos
Osteogênese , Coluna Vertebral , Ratos , Camundongos , Coelhos , Animais , Colágeno
2.
Biomaterials ; 301: 122238, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37441901

RESUMO

Spherical 50 nm silica-based nanoparticles (SiNPs) promote healthy bone homeostasis and maintenance by supporting bone forming osteoblast lineage cells while simultaneously inhibiting the differentiation of bone resorbing osteoclasts. Previous work demonstrated that an intraperitoneal injection of SiNPs in healthy mice - both young and old - increased bone density and quality, suggesting the possibility that SiNPs represent a dual action therapeutic. However, the underlying mechanisms governing the osteoclast response to SiNPs have yet to be fully explored and defined. Therefore, the goals of this study were to investigate the cellular and molecular mechanisms by which SiNPs inhibit osteoclastogenesis. SiNPs strongly inhibited RANKL-induced osteoclast differentiation within the first hours and concomitantly inhibited early transcriptional regulators such as Nfatc1. SiNPs simultaneously stimulated expression of autophagy related genes p62 and LC3ß dependent on ERK1/2 signaling pathway. Intriguingly, SiNPs were found to stimulate autophagosome formation while inhibiting the autophagic flux necessary for RANKL-stimulated osteoclast differentiation, resulting in the inhibition of both the canonical and non-canonical NF-κB signaling pathways and stabilizing TRAF3. These results suggest a model in which SiNPs inhibit osteoclastogenesis by inhibiting the autophagic machinery and RANKL-dependent functionality. This mechanism of action defines a novel therapeutic strategy for inhibiting osteoclastogenesis.


Assuntos
Reabsorção Óssea , Osteogênese , Animais , Camundongos , NF-kappa B/metabolismo , Reabsorção Óssea/tratamento farmacológico , Osteoclastos/metabolismo , Diferenciação Celular , Autofagia , Ligante RANK/metabolismo , Fatores de Transcrição NFATC/metabolismo
3.
JCI Insight ; 8(10)2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37079375

RESUMO

The intake of dietary phosphate far exceeds recommended levels; however, the long-term health consequences remain relatively unknown. Here, the chronic physiological response to sustained elevated and reduced dietary phosphate consumption was investigated in mice. Although serum phosphate levels were brought into homeostatic balance, the prolonged intake of a high-phosphate diet dramatically and negatively impacted bone volume; generated a sustained increase in the phosphate responsive circulating factors FGF23, PTH, osteopontin and osteocalcin; and produced a chronic low-grade inflammatory state in the BM, marked by increased numbers of T cells expressing IL-17a, RANKL, and TNF-α. In contrast, a low-phosphate diet preserved trabecular bone while increasing cortical bone volume over time, and it reduced inflammatory T cell populations. Cell-based studies identified a direct response of T cells to elevated extracellular phosphate. Neutralizing antibodies against proosteoclastic cytokines RANKL, TNF-α, and IL-17a blunted the high-phosphate diet-induced bone loss identifying bone resorption as a regulatory mechanism. Collectively, this study illuminates that habitual consumption of a high-phosphate diet in mice induces chronic inflammation in bone, even in the absence of elevated serum phosphate. Furthermore, the study supports the concept that a reduced phosphate diet may be a simple yet effective strategy to reduce inflammation and improve bone health during aging.


Assuntos
Reabsorção Óssea , Fósforo na Dieta , Camundongos , Animais , Interleucina-17 , Fator de Necrose Tumoral alfa , Linfócitos T , Citocinas , Inflamação , Fosfatos
4.
Biochem Pharmacol ; 183: 114305, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129806

RESUMO

Phosphorus, often in the form of inorganic phosphate (Pi), is critical to cellular function on many levels; it is required as an integral component of kinase signaling, in the formation and function of DNA and lipids, and energy metabolism in the form of ATP. Accordingly, crucial aspects of cell mitosis - such as DNA synthesis and ATP energy generation - elevate the cellular requirement for Pi, with rapidly dividing cells consuming increased levels. Mechanisms to sense, respond, acquire, accumulate, and potentially seek Pi have evolved to support highly proliferative cellular states such as injury and malignant transformation. As such, manipulating Pi availability to target rapidly dividing cells presents a novel strategy to reduce or prevent unrestrained cell growth. Currently, limited knowledge exists regarding how modulating Pi consumption by pre-cancerous cells might influence the initiation of aberrant growth during malignant transformation, and if reducing the bioavailability or suppressing Pi consumption by malignant cells could alter tumorigenesis. The concept of targeting Pi-regulated pathways and/or consumption by pre-cancerous or tumor cells represents a novel approach to cancer prevention and control, although current data remains insufficient as to rigorously assess the therapeutic value and physiological relevance of this strategy. With this review, we present a critical evaluation of the paradox of how an element critical to essential cellular functions can, when available in excess, influence and promote a cancer phenotype. Further, we conjecture how Pi manipulation could be utilized as a therapeutic intervention, either systemically or at the cell level, to ultimately suppress or treat cancer initiation and/or progression.


Assuntos
Carcinogênese/induzido quimicamente , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Fosfatos/efeitos adversos , Fósforo na Dieta/efeitos adversos , Animais , Carcinogênese/patologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/induzido quimicamente , Neoplasias/patologia , Fosfatos/administração & dosagem , Fósforo na Dieta/administração & dosagem
5.
J Nutr Biochem ; 80: 108374, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32278118

RESUMO

Phosphorus is a common additive used in food processing that is typically consumed in excess of the recommended daily allowance; however, our knowledge of its effects on health, in the context of normal renal function, is limited. Unlike phosphorus, calcium intake is generally less than recommended, and it has been hypothesized that the calcium to phosphorus ratio may be partly responsible for the proposed negative health consequences. Therefore, this study sought to determine the effects of increased phosphorus additive intake, in the context of high calcium consumption, on endocrine markers of mineral metabolism and cardiometabolic health. An outpatient feeding study was performed in which healthy adults were fed a run-in control diet for 2 weeks followed by a phosphorus additive enhanced diet with supplemental calcium to an approximate ratio of 1 (experimental diet) for 2 weeks. Blood and urine samples were collected, and participants had brachial flow-mediated dilatation measured, with analyses comparing follow-up measures to baseline. Two weeks of experimental diet increased serum fibroblast growth factor 23 concentrations but lowered body weight and serum leptin; however, other phosphorus responsive factors such as osteopontin and osteocalcin did not increase. A complementary study in male mice also demonstrated that the regulation of known dietary phosphorus responsive factors was mostly abrogated when dietary calcium was raised in parallel with phosphorus. In conclusion, the study identifies weight, leptin and insulin as responsive to dietary phosphorus and that certain aspects of the systemic phosphorus response are attenuated by a corresponding high calcium intake.


Assuntos
Cálcio da Dieta/administração & dosagem , Doenças Cardiovasculares/epidemiologia , Minerais/metabolismo , Fósforo na Dieta/administração & dosagem , Adulto , Animais , Biomarcadores/sangue , Peso Corporal/efeitos dos fármacos , Cálcio/sangue , Dieta , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Aditivos Alimentares/administração & dosagem , Humanos , Insulina/metabolismo , Leptina/sangue , Masculino , Camundongos , Osteocalcina/metabolismo , Osteopontina/metabolismo , Fósforo/sangue
6.
Acta Biomater ; 82: 184-196, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30326276

RESUMO

Silica based nanoparticles have been demonstrated to have intrinsic biologic activity towards the skeleton and to function by promoting the differentiation of bone forming osteoblasts while inhibiting the differentiation of bone resorbing osteoclasts. The excitement surrounding nanomedicine in part revolves around the almost unlimited possibilities for varying the physicochemical properties including size, composition, and surface charge. To date few studies have attempted to manipulate these characteristics in concert to optimize a complex biologic outcome. Towards this end, spherical silica nanoparticles of various sizes (50-450 nm), of different surface properties (OH, CO2H, NR4+, mNH2), and of different composition (silica, gold, and polystyrene) were synthesized and evaluated for biological activity toward skeletal cells. Osteoblast activity was most influenced by composition and size variables, whereas osteoclasts were most affected by surface property variation. The study also establishes nanoparticle mediated suppression of Nfatc1, a key transcriptional regulator for osteoclast differentiation, identifying a novel mechanism of action. Collectively, the study highlights how during the design of bioactive nanoparticles, it is vital to consider not only the myriad of physical properties that can be manipulated, but also that the characteristics of the target cell plays an equally integral role in determining biological outcome. STATEMENT OF SIGNIFICANCE: Silica nanomaterials represent a promising biomaterial for beneficial effects on bone mass and quality as well as regenerative tissue engineering and are currently being investigated for intrinsic bioactivity towards the primary cells responsible for skeletal homeostasis; osteoblasts and osteoclasts. The goal of the current study was to assess the physical properties of silica nanoparticles that impart intrinsic bioactivity by evaluating size, surface charge, and composition. Results reveal differential influences of the physical properties of nanoparticles towards osteoblasts and osteoclasts. This study provides new insights into the design of nanoparticles to specifically target different aspects of bone metabolism and highlights the opportunities provided by nanotechnology to modulate a range of cell specific biological responses for therapeutic benefit.


Assuntos
Diferenciação Celular , Nanopartículas/química , Osteoclastos/metabolismo , Osteócitos/metabolismo , Dióxido de Silício/química , Animais , Camundongos , Osteoclastos/citologia , Osteócitos/citologia , Tamanho da Partícula , Células RAW 264.7 , Propriedades de Superfície
7.
Ann N Y Acad Sci ; 1415(1): 21-33, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29500936

RESUMO

Activated lymphocytes promote inflammation and bone destruction in rheumatoid arthritis (RA), making T cells and B cells therapeutic targets. Indeed, pharmacological blockade of CD28 costimulation using CTLA-4Ig (abatacept), approved for amelioration of RA, renders T cells dormant (anergic). CTLA-4Ig also promotes bone accretion in healthy mice; surprisingly, however, this effect is driven exclusively through upregulation of bone formation, rather than anti-inflammatory effects on resorption. In the study presented here, we utilized T cell receptor ß gene and Wnt-10b gene knockout mice to investigate the roles of T cells and Wnt-10b in CTLA-4Ig-induced bone anabolism. Ablation of either T cells or Wnt-10b not only abolished CTLA-4Ig-induced bone anabolism but also, paradoxically, suppressed bone formation leading to bone loss. Stalled bone formation was accompanied by bone marrow stromal cell expression of the Wnt pathway inhibitor sclerostin. Our data suggest that an immunoskeletal pivot may promote or suppress bone formation, depending on the net outcome of CTLA-4Ig action directed independently on T cells and osteoblast-linage cells that counter Wnt-10b-induced bone anabolism, by secretion of sclerostin. While CTLA-4Ig action is tipped in favor of bone formation under physiological conditions, pathological immunodeficiency may lead to suppressed bone formation and skeletal damage.


Assuntos
Abatacepte/farmacologia , Anabolizantes/farmacologia , Osso e Ossos/efeitos dos fármacos , Glicoproteínas/metabolismo , Linfócitos T/efeitos dos fármacos , Proteínas Wnt/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antirreumáticos/farmacologia , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Antígenos CD28/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Microtomografia por Raio-X
8.
Int J Nanomedicine ; 12: 8699-8710, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29263664

RESUMO

To date, delivery of light-emitting diode (LED)-activated compounds to cells and tissue remains a challenge. Silica-based materials possess good biocompatibility and have advantages of control of size and shape. Fluorescent silica nanoparticles (NPs) have been synthesized and used for applications such as cell tracking and tumor identification. Here, we report the synthesis and optimization of fluorescent silica NPs, which incorporate a naphthalimide dye with triethoxysilanes that are excited by the blue LED wavelength (LEDex NPs). The NPs can be imaged in the 420-470 nm wavelength, demonstrate a high quantum yield, are stable in a range of pH, and are taken into the cells. Therefore, these NPs represent a novel imaging technology for biomedical applications.


Assuntos
Corantes Fluorescentes/química , Nanopartículas/química , Dióxido de Silício/química , Animais , Células da Medula Óssea , Corantes Fluorescentes/síntese química , Concentração de Íons de Hidrogênio , Camundongos , Naftalimidas/química , Tamanho da Partícula
9.
ACS Appl Mater Interfaces ; 9(45): 39185-39196, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29045789

RESUMO

Hydroxyapatite (HAp) is critical to health both as the main structural material of the skeleton and storage material of calcium and phosphate. Nanosized HAp (nHAp) is naturally produced by mineralizing cells during bone formation and remodeling and is the main constituent of the skeleton. As such, HAp is currently being investigated as a therapeutic biomaterial for orthopedic and dental purposes. Recent studies have suggested that extracellular nHAp can influence osteoblast lineage commitment and cell function through changes in gene expression; however, the mechanisms remain to be elucidated. Here, the cellular and molecular mechanism by which rod-shaped nHAp (10 × 100 nm) stimulates gene expression in preosteoblast bone marrow stromal cells was investigated. Electron microscopy detected a rapid and stable interaction of nHAp with the cell membrane, which correlated with a strong stimulation of the Erk1/2 signaling pathway. Results also identified the requirement of the Fgf receptor signaling and phosphate-transporters for nHAp regulated gene expression whereas a calcium-sensing receptor inhibitor had no effect. Collectively, the study uncovers novel signaling pathways and cellular events specifically stimulated by and required for the cellular response to free extracellular HAp. The results provide insight into the osteoblastic response to HAp relevant to functional mineralization and pathological calcification and could be used in the development of biomaterials for orthopedic purposes.


Assuntos
Nanoestruturas , Linhagem Celular , Durapatita , Expressão Gênica , Sistema de Sinalização das MAP Quinases , Osteoblastos , Osteogênese , Proteínas de Transporte de Fosfato , Receptores de Fatores de Crescimento de Fibroblastos
10.
J Clin Endocrinol Metab ; 100(11): 4264-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26323022

RESUMO

CONTEXT: Phosphorus-based food additives can substantially increase total phosphorus intake per day, but the effect of these additives on endocrine factors regulating bone and mineral metabolism is unclear. OBJECTIVE: This study aimed to examine the effect of phosphorus additives on markers of bone and mineral metabolism. Design and Setting, and Participants: This was a feeding study of 10 healthy individuals fed a diet providing ∼1000 mg of phosphorus/d using foods known to be free of phosphorus additives for 1 week (low-additive diet), immediately followed by a diet containing identical food items; however, the foods contained phosphorus additives (additive-enhanced diet). Parallel studies were conducted in animals fed low- (0.2%) and high- (1.8%) phosphorus diets for 5 or 15 weeks. MAIN OUTCOME MEASURES: The changes in markers of mineral metabolism after each diet period were measured. RESULTS: Participants were 32 ± 8 years old, 30% male, and 70% black. The measured phosphorus content of the additive-enhanced diet was 606 ± 125 mg higher than the low-additive diet (P < .001). After 1 week of the low-additive diet, consuming the additive-enhanced diet for 1 week significantly increased circulating fibroblast growth factor 23 (FGF23), osteopontin, and osteocalcin concentrations by 23, 10, and 11%, respectively, and decreased mean sclerostin concentrations (P < .05 for all). Similarly, high-phosphorus diets in mice significantly increased blood FGF23, osteopontin and osteocalcin, lowered sclerostin, and decreased bone mineral density (P < .05 for all). CONCLUSIONS: The enhanced phosphorus content of processed foods can disturb bone and mineral metabolism in humans. The results of the animal studies suggest that this may compromise bone health.


Assuntos
Osso e Ossos/metabolismo , Aditivos Alimentares/farmacologia , Minerais/metabolismo , Compostos de Fósforo/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Animais , Biomarcadores/metabolismo , Densidade Óssea/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/sangue , Osso e Ossos/efeitos dos fármacos , Dieta , Comportamento Alimentar , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Marcadores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Osteocalcina/sangue , Osteopontina/sangue , Adulto Jovem
11.
Biomaterials ; 65: 32-42, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26141836

RESUMO

Hydroxyapatite (HA) is the primary structural component of the skeleton and dentition. Under biological conditions, HA does not occur spontaneously and therefore must be actively synthesized by mineralizing cells such as osteoblasts. The mechanism(s) by which HA is actively synthesized by cells and deposited to create a mineralized matrix are not fully understood and the consequences of mineralization on cell function are even less well understood. HA can be chemically synthesized (HAp) and is therefore currently being investigated as a promising therapeutic biomaterial for use as a functional scaffold and implant coating for skeletal repair and dental applications. Here we investigated the biological effects of nano-HAp (10 × 100 nm) on the lineage commitment and differentiation of bone forming osteoblasts. Exposure of early stage differentiating osteoblasts resulted in dramatic and sustained changes in gene expression, both increased and decreased, whereas later stage osteoblasts were much less responsive. Analysis of the promoter region one of the most responsive genes, alkaline phosphatase, identified the stimulation of DNA methylation following cell exposure to nano-HAp. Collectively, the results reveal the novel epigenetic regulation of cell function by nano-HAp which has significant implication on lineage determination as well as identifying a novel potential therapeutic use of nanomaterials.


Assuntos
Materiais Biocompatíveis/farmacologia , Metilação de DNA/efeitos dos fármacos , Durapatita/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Fosfatase Alcalina/genética , Animais , Materiais Biocompatíveis/química , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Durapatita/química , Epigênese Genética/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Camundongos , Nanoestruturas/química , Nanoestruturas/ultraestrutura , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos
12.
Nanomedicine ; 11(4): 959-967, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25680544

RESUMO

We recently reported that in vitro, engineered 50nm spherical silica nanoparticles promote the differentiation and activity of bone building osteoblasts but suppress bone-resorbing osteoclasts. Furthermore, these nanoparticles promote bone accretion in young mice in vivo. We have now investigated the capacity of these nanoparticles to reverse bone loss in aged mice, a model of human senile osteoporosis. Aged mice received nanoparticles weekly and bone mineral density (BMD), bone structure, and bone turnover were quantified. Our data revealed a significant increase in BMD, bone volume, and biochemical markers of bone formation. Biochemical and histological examinations failed to identify any abnormalities caused by nanoparticle administration. Our studies demonstrate that silica nanoparticles effectively blunt and reverse age-associated bone loss in mice by a mechanism involving promotion of bone formation. The data suggest that osteogenic silica nanoparticles may be a safe and effective therapeutic for counteracting age-associated bone loss. FROM THE CLINICAL EDITOR: Osteoporosis poses a significant problem in the society. Based on their previous in-vitro findings, the authors' group investigated the effects of spherical silica nanoparticles in reversing bone loss in a mouse model of osteoporosis. The results showed that intra-peritoneal injections of silica nanoparticles could increase bone mineral density, with little observed toxic side effects. This novel method may prove important in future therapy for combating osteoporosis.


Assuntos
Nanopartículas/química , Osteoblastos , Osteoclastos , Osteogênese/efeitos dos fármacos , Osteoporose , Dióxido de Silício , Animais , Biomarcadores/metabolismo , Densidade Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Humanos , Camundongos , Osteoblastos/diagnóstico por imagem , Osteoblastos/metabolismo , Osteoclastos/diagnóstico por imagem , Osteoclastos/metabolismo , Osteoporose/diagnóstico por imagem , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Radiografia , Dióxido de Silício/química , Dióxido de Silício/farmacologia
13.
Mol Carcinog ; 54(9): 926-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24700685

RESUMO

Recent studies in both rodents and humans suggest that elevated serum phosphorus, in the context of normal renal function, potentiates, or exacerbates pathologies associates with cardiovascular disease, bone metabolism, and cancer. Our recent microarray studies identified the potent stimulation of pro-angiogenic genes such as forkhead box protein C2 (FOXC2), osteopontin, and Vegfα, among others in response to elevated inorganic phosphate (Pi). Increased angiogenesis and neovascularization are important events in tumor growth and the progression to malignancy and FOXC2 has recently been identified as a potential transcriptional regulator of these processes. In this study we addressed the possibility that a high Pi environment would increase the angiogenic potential of cancer cells through a mechanism requiring FOXC2. Our studies utilized lung and breast cancer cell lines in combination with the human umbilical vascular endothelial cell (HUVEC) vessel formation model to better understand the mechanism(s) by which a high Pi environment might alter cancer progression. Exposure of cancer cells to elevated Pi stimulated expression of FOXC2 and conditioned medium from the Pi-stimulated cancer cells stimulated migration and tube formation in the HUVEC model. Mechanistically, we define the requirement of FOXC2 for Pi-induced osteopontin (OPN) expression and secretion from cancer cells as necessary for the angiogenic response. These studies reveal for the first time that cancer cells grown in a high Pi environment promote migration of endothelial cells and tube formation and in so doing identify a novel potential therapeutic target to reduce tumor progression.


Assuntos
Neoplasias da Mama/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Neoplasias Pulmonares/metabolismo , Neovascularização Patológica/metabolismo , Osteopontina/genética , Fosfatos/metabolismo , Mama/irrigação sanguínea , Mama/metabolismo , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Feminino , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , Neovascularização Patológica/genética , Osteopontina/metabolismo , Microambiente Tumoral
14.
ACS Nano ; 8(6): 5898-910, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24806912

RESUMO

We recently identified an engineered bioactive silica-based nanoparticle formulation (designated herein as NP1) that stimulates in vitro differentiation and mineralization of osteoblasts, the cells responsible for bone formation, and increases bone mineral density in young mice in vivo. The results demonstrate that these nanoparticles have intrinsic biological activity; however, the intracellular fate and a complete understanding of the mechanism(s) involved remains to be elucidated. Here we investigated the cellular mechanism(s) by which NP1 stimulates differentiation and mineralization of osteoblasts. We show that NP1 enters the cells through a caveolae-mediated endocytosis followed by stimulation of the mitogen activated protein kinase ERK1/2 (p44/p42). Our findings further revealed that NP1 stimulates autophagy including the processing of LC3ß-I to LC3ß-II, a key protein involved in autophagosome formation, which is dependent on ERK1/2 signaling. Using a variant of NP1 with cobalt ferrite magnetic metal core (NP1-MNP) to pull down associated proteins, we found direct binding of LC3ß and p62, two key proteins involved in autophagosome formation, with silica nanoparticles. Interestingly, NP1 specifically interacts with the active and autophagosome associated form of LC3ß (LC3ß-II). Taken together, the stimulation of autophagy and associated signaling suggests a cellular mechanism for the stimulatory effects of silica nanoparticles on osteoblast differentiation and mineralization.


Assuntos
Nanopartículas Metálicas/química , Proteínas Associadas aos Microtúbulos/metabolismo , Osteoblastos/citologia , Proteínas de Ligação a RNA/metabolismo , Dióxido de Silício/química , Autofagia , Diferenciação Celular , Endocitose , Endossomos/metabolismo , Humanos , Lisossomos/metabolismo , Microscopia de Fluorescência , Nanotecnologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fagossomos/metabolismo , Ligação Proteica
16.
Toxicol In Vitro ; 28(3): 354-64, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24333519

RESUMO

Silica-based nanomaterials are generally considered to be excellent candidates for therapeutic applications particularly related to skeletal metabolism however the current data surrounding the safety of silica based nanomaterials is conflicting. This may be due to differences in size, shape, incorporation of composite materials, surface properties, as well as the presence of contaminants following synthesis. In this study we performed extensive in vitro safety profiling of ∼ 50 nm spherical silica nanoparticles with OH-terminated or Polyethylene Glycol decorated surface, with and without a magnetic core, and synthesized by the Stöber method. Nineteen different cell lines representing all major organ types were used to investigate an in vitro lethal concentration (LC) and results revealed little toxicity in any cell type analyzed. To calculate an in vitro therapeutic index we quantified the effective concentration at 50% response (EC50) for nanoparticle-stimulated mineral deposition activity using primary bone marrow stromal cells (BMSCs). The EC50 for BMSCs was not substantially altered by surface or magnetic core. The calculated Inhibitory concentration 50% (IC50) for pre-osteoclasts was similar to the osteoblastic cells. These results demonstrate the pharmacological potential of certain silica-based nanomaterial formulations for use in treating bone diseases based on a favorable in vitro therapeutic index.


Assuntos
Magnetismo , Nanopartículas , Polietilenoglicóis/química , Dióxido de Silício/farmacologia , Animais , Doenças Ósseas/tratamento farmacológico , Doenças Ósseas/patologia , Linhagem Celular , Humanos , Concentração Inibidora 50 , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Tamanho da Partícula , Dióxido de Silício/administração & dosagem , Dióxido de Silício/toxicidade , Testes de Toxicidade/métodos
17.
PLoS One ; 8(10): e77121, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24194864

RESUMO

The sodium-dependent phosphate co-transporter 2b (NPT2b) plays an important role in maintaining phosphate homeostasis. In previous studies, we have shown that high dietary inorganic phosphate (Pi) consumption in mice stimulated lung tumorigenesis and increased NPT2b expression. NPT2b has also been found to be highly expressed in human lung cancer tissues. The association of high expression of NPT2b in the lung with poor prognosis in oncogenic lung diseases prompted us to test whether knockdown of NPT2b may regulate lung cancer growth. To address this issue, aerosols that contained small interfering RNA (siRNA) directed against NPT2b (siNPT2b) were delivered into the lungs of K-ras (LA1) mice, which constitute a murine model reflecting human lung cancer. Our results clearly showed that repeated aerosol delivery of siNPT2b successfully suppressed lung cancer growth and decreased cancer cell proliferation and angiogenesis, while facilitating apoptosis. These results strongly suggest that NPT2b plays a role lung tumorigenesis and represents a novel target for lung cancer therapy.


Assuntos
Carcinogênese/genética , Neoplasias Pulmonares/prevenção & controle , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Aerossóis/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Carcinogênese/efeitos dos fármacos , Primers do DNA/genética , Técnicas de Silenciamento de Genes/métodos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Neoplasias Pulmonares/genética , Camundongos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real
18.
Soft Mater ; 11(2): 195-203, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24058289

RESUMO

Nanomaterials are diverse in size, shape and charge and these differences likely alter their physicochemical properties in biological systems. We have investigated how these properties alter the initial and long-term dynamics of endocytosis, cell viability, cell division, exocytosis, and interaction with a collagen extracellular matrix using silica-based fluorescent nanoparticles and the murine pre-osteoblast cell line, MC3T3-E1. Three surface modified nanoparticles were analyzed: positively charged (PTMA), negatively charged (OH), and neutrally charged polyethylene glycol (PEG). Positively charged PTMA-modified nanoparticles demonstrated the most rapid uptake, within 2 hours, while PEG modified and negatively charged OH nanoparticles demonstrated slower uptake. Cell viability was >80% irrespective of nanoparticle surface charge suggesting a general lack of toxicity. Long-term monitoring of fluorescent intensity revealed that nanoparticles were passed to daughter cells during mitotic cell division with a corresponding decrease in fluorescent intensity. These data suggest that irrespective of surface charge silica nanoparticles have the potential to internalize into osteoblasts, albeit with different kinetics. Furthermore, long lived nanoparticles have the potential to be transferred to daughter cells during mitosis and can be maintained for weeks intracellularly or within a collagen matrix without toxicity and limited exocytosis.

19.
J Cell Physiol ; 228(7): 1536-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23280476

RESUMO

Recent studies have suggested that changes in serum phosphate levels influence pathological states associated with aging such as cancer, bone metabolism, and cardiovascular function, even in individuals with normal renal function. The causes are only beginning to be elucidated but are likely a combination of endocrine, paracrine, autocrine, and cell autonomous effects. We have used an integrated quantitative biology approach, combining transcriptomics and proteomics to define a multi-phase, extracellular phosphate-induced, signaling network in pre-osteoblasts as well as primary human and mouse mesenchymal stromal cells. We identified a rapid mitogenic response stimulated by elevated phosphate that results in the induction of immediate early genes including c-fos. The mechanism of activation requires FGF receptor signaling followed by stimulation of N-Ras and activation of AP-1 and serum response elements. A distinct long-term response also requires FGF receptor signaling and results in N-Ras activation and expression of genes and secretion of proteins involved in matrix regulation, calcification, and angiogenesis. The late response is synergistically enhanced by addition of FGF23 peptide. The intermediate phase results in increased oxidative phosphorylation and ATP production and is necessary for the late response providing a functional link between the phases. Collectively, the results define elevated phosphate, as a mitogen and define specific mechanisms by which phosphate stimulates proliferation and matrix regulation. Our approach provides a comprehensive understanding of the cellular response to elevated extracellular phosphate, functionally connecting temporally coordinated signaling, transcriptional, and metabolic events with changes in long-term cell behavior.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fosfatos/metabolismo , Transdução de Sinais/fisiologia , Células 3T3 , Trifosfato de Adenosina/biossíntese , Animais , Células Cultivadas , Biologia Computacional , Espaço Extracelular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Genes Precoces , Genes fos , Genes ras , Humanos , Camundongos , Neovascularização Fisiológica , Osteoblastos/metabolismo , Regiões Promotoras Genéticas , Proteínas/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo
20.
Cell Oncol (Dordr) ; 36(1): 15-26, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23070870

RESUMO

BACKGROUND: Osteopontin (OPN) is a secreted glycophosphoprotein that has been implicated in the regulation of cancer development. The function of OPN is primarily regulated through post-translational modification such as glycosylation. As yet, however, the relationship between OPN glycosylation and lung cancer development has not been investigated. In this study, we addressed this issue by studying the effect of a triple mutant (TM) of OPN, which is mutated at three O-glycosylation sites, on lung cancer development in K-ras (LA1) mice, a murine model for human non-small cell lung cancer. METHODS: Aerosolized lentivirus-based OPN TM was delivered into the lungs of K-ras (LA1) mice using a nose-only-inhalation chamber 3 times/wk for 4 wks. Subsequently, the effects of repeated delivery of OPN TM on lung tumorigenesis and its concomitant OPN-mediated signaling pathways were investigated. RESULTS: Aerosol-delivered OPN TM inhibited lung tumorigenesis. In addition, the OPN-mediated Akt signaling pathway was inhibited. OPN TM also decreased NF-κB activity and the phosphorylation of 4E-BP1, while facilitating apoptosis in the lungs of K-ras (LA1) mice. CONCLUSIONS: Our results show that aerosol delivery of OPN TM successfully suppresses lung cancer development in the K-ras (LA1) mouse model and, therefore, warrant its further investigation as a possible therapeutic strategy for non-small cell lung cancer.


Assuntos
Neoplasias Pulmonares/terapia , Mutação , Osteopontina/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Adaptadoras de Transdução de Sinal , Aerossóis/administração & dosagem , Animais , Apoptose/genética , Western Blotting , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Fatores de Iniciação em Eucariotos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glicosilação , Lentivirus/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , NF-kappa B/metabolismo , Osteopontina/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Carga Tumoral/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...