Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Genet ; 13(8): e1006975, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28827794

RESUMO

DJ-1 is one of the causative genes for early onset familiar Parkinson's disease (PD) and is also considered to influence the pathogenesis of sporadic PD. DJ-1 has various physiological functions which converge on controlling intracellular reactive oxygen species (ROS) levels. In RNA-sequencing analyses searching for novel anti-oxidant genes downstream of DJ-1, a gene encoding NADP+-dependent isocitrate dehydrogenase (IDH), which converts isocitrate into α-ketoglutarate, was detected. Loss of IDH induced hyper-sensitivity to oxidative stress accompanying age-dependent mitochondrial defects and dopaminergic (DA) neuron degeneration in Drosophila, indicating its critical roles in maintaining mitochondrial integrity and DA neuron survival. Further genetic analysis suggested that DJ-1 controls IDH gene expression through nuclear factor-E2-related factor2 (Nrf2). Using Drosophila and mammalian DA models, we found that IDH suppresses intracellular and mitochondrial ROS level and subsequent DA neuron loss downstream of DJ-1. Consistently, trimethyl isocitrate (TIC), a cell permeable isocitrate, protected mammalian DJ-1 null DA cells from oxidative stress in an IDH-dependent manner. These results suggest that isocitrate and its derivatives are novel treatments for PD associated with DJ-1 dysfunction.


Assuntos
Proteínas de Drosophila/genética , Isocitrato Desidrogenase/genética , Degeneração Neural/genética , Proteínas do Tecido Nervoso/genética , Doença de Parkinson/genética , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Isocitratos/metabolismo , Mitocôndrias/genética , Mitocôndrias/patologia , NADP/genética , Fator de Transcrição NF-E2/genética , Degeneração Neural/fisiopatologia , Estresse Oxidativo/genética , Doença de Parkinson/patologia
2.
Biochem Biophys Res Commun ; 508(1): 308-313, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30497781

RESUMO

Wnt signaling pathway plays critical roles in body axes patterning, cell fate specification, cell proliferation, cell migration, stem cell maintenance, cancer development and etc. Deregulation of this pathway can be causative of cancer, metabolic disease and neurodegenerative disease such as Parkinson`s disease. Among the core components of Wnt signaling pathway, we discovered that Dishevelled (Dsh) interacts with ULK1 and is phosphorylated by ULK1. Unexpectedly, the knockdown of ULK1 elicited a marked increase in Wnt/ß-catenin signaling. Multiple ULK1 phosphorylation sites existed on Dsh and many of them were located on the PDZ-DEP region. By using evolutionarily well conserved Drosophila Dsh, we found that S239, S247 and S254 in the PDZ-DEP region are involved in phosphorylation of Dsh by ULK1. Among these, S247 and S254 were conserved in human Dsh. When phospho-mimetic mutants (2D and 2E Dsh mutants) of these conserved residues were generated and expressed in the eyes of the fruit flies, the activity of Dsh was significantly decreased compared to wild type Dsh. Through additional alanine scanning, we further identified that S239, S247, S254, S266, S376, S554 and S555 on full length Dsh were phosphorylated by ULK1. In regards to the S266A mutation located in the PDZ domain among these phosphorylated residues, our results suggested that Dsh forms an SDS-resistant high molecular weight complex with ß-catenin and TCF in the nucleus in an S266 phosphorylation-dependent manner. Based on these results, we propose that ULK1 plays a pivotal role in the regulation of Wnt/ß-catenin signaling pathway by phosphorylating Dsh.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteínas Desgrenhadas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Via de Sinalização Wnt , Células Cultivadas , Células HEK293 , Humanos , Fosforilação
3.
Molecules ; 19(8): 12727-59, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25140450

RESUMO

Chronic kidney disease (CKD) is featured by a progressive decline of kidney function and is mainly caused by chronic diseases such as diabetes mellitus and hypertension. CKD is a complex disease due to cardiovascular complications and high morbidity; however, there is no single treatment to improve kidney function in CKD patients. Since biological markers representing oxidative stress are significantly elevated in CKD patients, oxidative stress is receiving attention as a contributing factor to CKD pathology. Nuclear factor erythroid-2 related factor 2 (NRF2) is a predominant transcription factor that regulates the expression of a wide array of genes encoding antioxidant proteins, thiol molecules and their generating enzymes, detoxifying enzymes, and stress response proteins, all of which can counteract inflammatory and oxidative damages. There is considerable experimental evidence suggesting that NRF2 signaling plays a protective role in renal injuries that are caused by various pathologic conditions. In addition, impaired NRF2 activity and consequent target gene repression have been observed in CKD animals. Therefore, a pharmacological intervention activating NRF2 signaling can be beneficial in protecting against kidney dysfunction in CKD. This review article provides an overview of the role of NRF2 in experimental CKD models and describes current findings on the renoprotective effects of naturally occurring NRF2 activators, including sulforaphane, resveratrol, curcumin, and cinnamic aldehyde. These experimental results, coupled with recent clinical experiences with a synthetic triterpenoid, bardoxolone methyl, have brought a light of hope for ameliorating CKD progression by preventing oxidative stress and maintaining cellular redox homeostasis.


Assuntos
Antioxidantes/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Oxirredução , Insuficiência Renal Crônica/genética , Animais , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/patologia , Masculino , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/genética , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais/genética
4.
J Biomech Eng ; 2013 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-23897168

RESUMO

Biophysical strain has been applied widely for bone regeneration. However, application of low-magnitude strains to cells on small-thickness scaffolds is problematic, especially in rodent calvarial defect models, because general translation systems have limitations in terms of generating low-magnitude smooth signals. To overcome these limitations, we developed an in vitro biophysical-stimulation platform for stimulation of cells on small-thickness scaffolds for rodent calvarial bone defects. The customized flexure-based translational nanoactuator enables generation of low-magnitude smooth signals at the subnano- to micrometer-scale. This nanoactuator, which is equipped with a piezoelectric actuator, is suitable for biological applications because it can generate friction-free motion with a high resolution. Moreover, its operation without wear or deterioration eliminates contamination factors in cell culture environments. The developed in vitro biophysical-stimulation platform using these nanoactuators showed predictable operational characteristics. Also, a few-micrometer sinusoidal signal was generated successfully without any distortion. Three-dimensional scaffolds fitting the critical-size rat calvarial defect model were fabricated using poly(caprolactone), poly(lactic-co-glycolic acid), and tricalcium phosphate. Runt-related transcription factor 2 expression was increased upon stimulation of human adipose-derived stem cells (ASCs) on these scaffolds were stimulated in the in vitro biophysical-stimulation platform. Additionally, the use of this platform resulted in up-regulation of alkaline phosphate, osteopontin, and osterix expression compared to the non-stimulated group. These preliminary in vitro results suggest that the biophysical environment provided by the in vitro biophysical-stimulation platform influences the osteogenic differentiation of ASCs.

5.
J Ultrasound Med ; 32(8): 1461-70, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23887957

RESUMO

OBJECTIVES: The purpose of this study was to demonstrate that the effects of continuous ultrasound on the osteogenic differentiation of human adipose-derived stem cells (hASCs) are dependent on the frequency in vitro. METHODS: Before stimulation, we characterized the hASCs using cluster of differentiation marker profiles and tridifferentiation. Then we selected effective frequencies in the range of 0.5 to 1.5 MHz (with a peak negative pressure of 52 kPa), which upregulated runt-related transcription factor 2 messenger RNA expression. Next, the effects of ultrasound at the selected frequencies on the osteogenic differentiation were evaluated at the protein level. Alkaline phosphatase activity and the formation of mineralized nodules were measured. We additionally identified the cellular mechanisms underlying the effects of ultrasound stimulation using Western blotting. RESULTS: The hASCs showed general cluster of differentiation marker profiles of stem cells and confirmed their potentials to yield adipogenic, chondrogenic, and osteogenic differentiation. Frequencies of 0.5, 1.0, and 1.5 MHz were selected for higher runt-related transcription factor 2 expression in the range of 0.5 to 1.5 MHz. Among the 3 groups, alkaline phosphatase activity and the formation of mineralized nodules were increased in cells exposed to 1.5-MHz ultrasound compared with cells exposed to 0.5-or 1.0-MHz ultrasound and nontreated control cells. We additionally confirmed that this acceleration of osteogenic differentiation was related to p38 and protein kinase B signaling pathways. CONCLUSIONS: In this study, we found that, in the selected range, 1.5 MHz was the most effective frequency for inducing the osteogenic differentiation of hASCs.


Assuntos
Adipócitos/citologia , Adipócitos/fisiologia , Osteoblastos/citologia , Osteoblastos/fisiologia , Sonicação/métodos , Células-Tronco/citologia , Células-Tronco/fisiologia , Adipócitos/efeitos da radiação , Diferenciação Celular/fisiologia , Diferenciação Celular/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Humanos , Osteoblastos/efeitos da radiação , Osteogênese/fisiologia , Osteogênese/efeitos da radiação , Doses de Radiação
6.
Nat Cancer ; 3(8): 994-1011, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35788723

RESUMO

We analyzed the contributions of structural variants (SVs) to gliomagenesis across 179 pediatric high-grade gliomas (pHGGs). The most recurrent SVs targeted MYC isoforms and receptor tyrosine kinases (RTKs), including an SV amplifying a MYC enhancer in 12% of diffuse midline gliomas (DMG), indicating an underappreciated role for MYC in pHGG. SV signature analysis revealed that tumors with simple signatures were TP53 wild type (TP53WT) but showed alterations in TP53 pathway members PPM1D and MDM4. Complex signatures were associated with direct aberrations in TP53, CDKN2A and RB1 early in tumor evolution and with later-occurring extrachromosomal amplicons. All pHGGs exhibited at least one simple-SV signature, but complex-SV signatures were primarily restricted to subsets of H3.3K27M DMGs and hemispheric pHGGs. Importantly, DMGs with complex-SV signatures were associated with shorter overall survival independent of histone mutation and TP53 status. These data provide insight into the impact of SVs on gliomagenesis and the mechanisms that shape them.


Assuntos
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/genética , Criança , Glioma/genética , Histonas/genética , Humanos , Mutação , Proteínas Proto-Oncogênicas/genética
7.
Bone ; 120: 166-175, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30409757

RESUMO

High-bone-mass (HBM)-causing missense mutations in the low density lipoprotein receptor-related protein-5 (Lrp5) are associated with increased osteoanabolic action and protection from disuse- and ovariectomy-induced osteopenia. These mutations (e.g., A214V and G171V) confer resistance to endogenous secreted Lrp5/6 inhibitors, such as sclerostin (SOST) and Dickkopf homolog-1 (DKK1). Cells in the osteoblast lineage are responsive to canonical Wnt stimulation, but recent work has indicated that osteoclasts exhibit both indirect and direct responsiveness to canonical Wnt. Whether Lrp5-HBM receptors, expressed in osteoclasts, might alter osteoclast differentiation, activity, and consequent net bone balance in the skeleton, is not known. To address this, we bred mice harboring heterozygous Lrp5 HBM-causing conditional knock-in alleles to Ctsk-Cre transgenic mice and studied the phenotype using DXA, µCT, histomorphometry, serum assays, and primary cell culture. Mice with HBM alleles induced in Ctsk-expressing cells (TG) exhibited higher bone mass and architectural properties compared to non-transgenic (NTG) counterparts. In vivo and in vitro measurements of osteoclast activity, population density, and differentiation yielded significant reductions in osteoclast-related parameters in female but not male TG mice. Droplet digital PCR performed on osteocyte enriched cortical bone tubes from TG and NTG mice revealed that ~8-17% of the osteocyte population (depending on sex) underwent recombination of the conditional Lrp5 allele in the presence of Ctsk-Cre. Further, bone formation parameters in the midshaft femur cortex show a small but significant increase in anabolic action on the endocortical but not periosteal surface. These findings suggest that Wnt/Lrp5 signaling in osteoclasts affects osteoclastogenesis and activity in female mice, but also that some of the changes in bone mass in TG mice might be due to Cre expression in the osteocyte population.


Assuntos
Osso e Ossos/metabolismo , Catepsina K/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação/genética , Absorciometria de Fóton , Alelos , Animais , Biomarcadores/sangue , Células da Medula Óssea/metabolismo , Reabsorção Óssea/sangue , Reabsorção Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Diferenciação Celular , Feminino , Integrases/metabolismo , Masculino , Camundongos Transgênicos , Tamanho do Órgão/genética , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/genética , Periósteo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Transgenes , Microtomografia por Raio-X
8.
Nat Biomed Eng ; 3(7): 509-519, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31148598

RESUMO

Patient-specific ex vivo models of human tumours that recapitulate the pathological characteristics and complex ecology of native tumours could help determine the most appropriate cancer treatment for individual patients. Here, we show that bioprinted reconstituted glioblastoma tumours consisting of patient-derived tumour cells, vascular endothelial cells and decellularized extracellular matrix from brain tissue in a compartmentalized cancer-stroma concentric-ring structure that sustains a radial oxygen gradient, recapitulate the structural, biochemical and biophysical properties of the native tumours. We also show that the glioblastoma-on-a-chip reproduces clinically observed patient-specific resistances to treatment with concurrent chemoradiation and temozolomide, and that the model can be used to determine drug combinations associated with superior tumour killing. The patient-specific tumour-on-a-chip model might be useful for the identification of effective treatments for glioblastoma patients resistant to the standard first-line treatment.


Assuntos
Bioimpressão/métodos , Quimiorradioterapia/métodos , Glioblastoma/tratamento farmacológico , Dispositivos Lab-On-A-Chip , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Combinação de Medicamentos , Avaliação de Medicamentos , Sinergismo Farmacológico , Células Endoteliais , Regulação Neoplásica da Expressão Gênica , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Oxigênio , Temozolomida/farmacologia , Microambiente Tumoral/efeitos dos fármacos
9.
J Neurosurg ; 129(4): 1085-1091, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29125417

RESUMO

OBJECTIVE: Cranial defects can result from trauma, infection, congenital malformations, and iatrogenic causes and represent a surgical challenge. The current standard of care is cranioplasty, with either autologous or allogeneic material. In either case, the intrinsic vascularity of the surrounding tissues allows for bone healing. The objective of this study was to determine if mechanotransductive gene manipulation would yield non-weight-bearing bone regeneration in a critical size calvarial defect in mice. METHODS: A mouse model of Sost deletion in Sost knockout (KO) mice was created in which the osteocytes do not express sclerostin. A critical size calvarial defect (4 mm in diameter) was surgically created in the parietal bone in 8-week-old wild-type (n = 8) and Sost KO (n = 8) male mice. The defects were left undisturbed (no implant or scaffold) to simulate a traumatic calvariectomy model. Eight weeks later, the animals were examined at necropsy by planimetry, histological analysis of new bone growth, and micro-CT scanning of bone thickness. RESULTS: Defects created in wild-type mice did not fill with bone over the study period of 2 months. Genetic downregulation of sclerostin yielded animals that were able to regenerate 40% of the initial critical size defect area 8 weeks after surgery. A thin layer of bone covered a significant portion of the original defect in all Sost KO animals. A statistically significant increase in bone volume (p < 0.05) was measured in Sost KO mice using radiodensitometric analysis. Immunohistochemical analysis also confirmed that this bone regeneration occurred through the Wnt pathway and originated from the edge of the defect; BMP signaling did not appear to be affected by sclerostin. CONCLUSIONS: Mechanical loading is an important mechanism of bone formation in the cranial skeleton and is poorly understood. This is partially due to the fact that it is difficult to load bone in the craniomaxillofacial skeleton. This study suggests that modulation of the Wnt pathway, as is able to be done with monoclonal antibodies, is a potentially efficacious method for bone regeneration that requires further study.


Assuntos
Desenvolvimento Ósseo/genética , Regeneração Óssea/genética , Deleção Cromossômica , Modelos Animais de Doenças , Glicoproteínas/genética , Mecanotransdução Celular/genética , Crânio/crescimento & desenvolvimento , Proteínas Adaptadoras de Transdução de Sinal , Animais , Regulação para Baixo/genética , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Osteócitos/fisiologia , Microtomografia por Raio-X
10.
JCI Insight ; 3(11)2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29875318

RESUMO

The WNT pathway has become an attractive target for skeletal therapies. High-bone-mass phenotypes in patients with loss-of-function mutations in the LRP5/6 inhibitor Sost (sclerosteosis), or in its downstream enhancer region (van Buchem disease), highlight the utility of targeting Sost/sclerostin to improve bone properties. Sclerostin-neutralizing antibody is highly osteoanabolic in animal models and in human clinical trials, but antibody-based inhibition of another potent LRP5/6 antagonist, Dkk1, is largely inefficacious for building bone in the unperturbed adult skeleton. Here, we show that conditional deletion of Dkk1 from bone also has negligible effects on bone mass. Dkk1 inhibition increases Sost expression, suggesting a potential compensatory mechanism that might explain why Dkk1 suppression lacks anabolic action. To test this concept, we deleted Sost from osteocytes in, or administered sclerostin neutralizing antibody to, mice with a Dkk1-deficient skeleton. A robust anabolic response to Dkk1 deletion was manifest only when Sost/sclerostin was impaired. Whole-body DXA scans, µCT measurements of the femur and spine, histomorphometric measures of femoral bone formation rates, and biomechanical properties of whole bones confirmed the anabolic potential of Dkk1 inhibition in the absence of sclerostin. Further, combined administration of sclerostin and Dkk1 antibody in WT mice produced a synergistic effect on bone gain that greatly exceeded individual or additive effects of the therapies, confirming the therapeutic potential of inhibiting multiple WNT antagonists for skeletal health. In conclusion, the osteoanabolic effects of Dkk1 inhibition can be realized if sclerostin upregulation is prevented. Anabolic therapies for patients with low bone mass might benefit from a strategy that accounts for the compensatory milieu of WNT inhibitors in bone tissue.


Assuntos
Anabolizantes/administração & dosagem , Glicoproteínas/antagonistas & inibidores , Hiperostose/tratamento farmacológico , Osteogênese/efeitos dos fármacos , Sindactilia/tratamento farmacológico , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos Neutralizantes/administração & dosagem , Proteínas Morfogenéticas Ósseas/genética , Modelos Animais de Doenças , Feminino , Fêmur/citologia , Fêmur/diagnóstico por imagem , Fêmur/patologia , Marcadores Genéticos/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Hiperostose/diagnóstico por imagem , Hiperostose/genética , Hiperostose/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mutação com Perda de Função , Masculino , Camundongos , Osteócitos , Coluna Vertebral/citologia , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/patologia , Sindactilia/diagnóstico por imagem , Sindactilia/genética , Sindactilia/patologia , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Microtomografia por Raio-X
11.
J Bone Miner Res ; 33(5): 930-944, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29314250

RESUMO

Approximately 10% of all bone fractures do not heal, resulting in patient morbidity and healthcare costs. However, no pharmacological treatments are currently available to promote efficient bone healing. Inhibition of Ca2+ /calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) reverses age-associated loss of trabecular and cortical bone volume and strength in mice. In the current study, we investigated the role of CaMKK2 in bone fracture healing and show that its pharmacological inhibition using STO-609 accelerates early cellular and molecular events associated with endochondral ossification, resulting in a more rapid and efficient healing of the fracture. Within 7 days postfracture, treatment with STO-609 resulted in enhanced Indian hedgehog signaling, paired-related homeobox (PRX1)-positive mesenchymal stem cell (MSC) recruitment, and chondrocyte differentiation and hypertrophy, along with elevated expression of osterix, vascular endothelial growth factor, and type 1 collagen at the fracture callus. Early deposition of primary bone by osteoblasts resulted in STO-609-treated mice possessing significantly higher callus bone volume by 14 days following fracture. Subsequent rapid maturation of the bone matrix bestowed fractured bones in STO-609-treated animals with significantly higher torsional strength and stiffness by 28 days postinjury, indicating accelerated healing of the fracture. Previous studies indicate that fixed and closed femoral fractures in the mice take 35 days to fully heal without treatment. Therefore, our data suggest that STO-609 potentiates a 20% acceleration of the bone healing process. Moreover, inhibiting CaMKK2 also imparted higher mechanical strength and stiffness at the contralateral cortical bone within 4 weeks of treatment. Taken together, the data presented here underscore the therapeutic potential of targeting CaMKK2 to promote efficacious and rapid healing of bone fractures and as a mechanism to strengthen normal bones. © 2018 American Society for Bone and Mineral Research.


Assuntos
Calo Ósseo/enzimologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Consolidação da Fratura/fisiologia , Proteínas Hedgehog/metabolismo , Osteogênese/fisiologia , Transdução de Sinais/fisiologia , Animais , Benzimidazóis/farmacologia , Colágeno Tipo I/metabolismo , Consolidação da Fratura/efeitos dos fármacos , Masculino , Camundongos , Naftalimidas/farmacologia , Osteogênese/efeitos dos fármacos
12.
Bone ; 88: 138-145, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27143110

RESUMO

Mechanical signal transduction in bone tissue begins with load-induced activation of several cellular pathways in the osteocyte population. A key pathway that participates in mechanotransduction is Wnt/Lrp5 signaling. A putative downstream mediator of activated Lrp5 is the nucleocytoplasmic shuttling protein ß-catenin (ßcat), which migrates to the nucleus where it functions as a transcriptional co-activator. We investigated whether osteocytic ßcat participates in Wnt/Lrp5-mediated mechanotransduction by conducting ulnar loading experiments in mice with or without chemically induced ßcat deletion in osteocytes. Mice harboring ßcat floxed loss-of-function alleles (ßcat(f/f)) were bred to the inducible osteocyte Cre transgenic (10)(kb)Dmp1-CreERt2. Adult male mice were induced to recombine the ßcat alleles using tamoxifen, and intermittent ulnar loading sessions were applied over the following week. Although adult-onset deletion of ßcat from Dmp1-expressing cells reduced skeletal mass, the bone tissue was responsive to mechanical stimulation as indicated by increased relative periosteal bone formation rates in recombined mice. However, load-induced improvements in cross sectional geometric properties were compromised in recombined mice. The collective results indicate that the osteoanabolic response to loading can occur on the periosteal surface when ß-cat levels are significantly reduced in Dmp1-expressing cells, suggesting that either (i) only low levels of ß-cat are required for mechanically induced bone formation on the periosteal surface, or (ii) other additional downstream mediators of Lrp5 might participate in transducing load-induced Wnt signaling.


Assuntos
Adaptação Fisiológica , Proteínas da Matriz Extracelular/metabolismo , Deleção de Genes , Osteoblastos/metabolismo , Osteócitos/metabolismo , Osteogênese , Periósteo/fisiopatologia , beta Catenina/metabolismo , Absorciometria de Fóton , Alelos , Animais , Animais Recém-Nascidos , Densidade Óssea , Osso Cortical/patologia , Camundongos , Músculos/patologia , Periósteo/patologia , Transgenes , Suporte de Carga
13.
Endocrinology ; 157(8): 3047-57, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27253995

RESUMO

ß-Catenin (ßcat) is a major downstream signaling node in canonical Wingless-related integration site (Wnt) signaling pathway, and its activity is crucial for canonical Wnt signal transduction. Wnt signaling has recently been implicated in the osteo-anabolic response to PTH, a potent calcium-regulating factor. We investigated whether ßcat is essential for the anabolic action of intermittent PTH by generating male mice with adult-onset deletion of ßcat in a subpopulation of bone cells (osteocytes and late-stage osteoblasts), treating them with an anabolic regimen of PTH, and measuring the skeletal responses. Male (10kb)Dmp1-CreERt2 transgenic mice that also harbored floxed loss-of-function ßcat alleles (ßcat(f/f)) were induced for Cre activity using tamoxifen, then injected daily with human PTH 1-34 (30 µg/kg) or vehicle for 5 weeks. Mice in which ßcat was deleted showed either total lack of bone mineral density (BMD) gain, or BMD loss, and did not respond to PTH treatment. However, bone mass measurements in the trabecular compartment of the femur and spine revealed PTH-induced bone gain whether ßcat was deleted or not. PTH-stimulated increases in periosteal and cancellous bone formation rates were not impaired by ßcat deletion, but resorption markers and cortical porosity were significantly increased in induced mice, particularly induced mice treated with PTH. These results suggest that ßcat is required for net-positive BMD effects of PTH therapy but that the anabolic effects per se of PTH treatment might not require osteocytic/osteoblastic ßcat.


Assuntos
Proteínas da Matriz Extracelular/genética , Deleção de Genes , Osteogênese , Hormônio Paratireóideo/farmacologia , beta Catenina/genética , Fatores Etários , Anabolizantes/metabolismo , Anabolizantes/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Hormônio Paratireóideo/metabolismo
14.
Biofabrication ; 8(1): 015007, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26756962

RESUMO

Several studies have focused on the regeneration of liver tissue in a two-dimensional (2D) planar environment, whereas actual liver tissue is three-dimensional (3D). Cell printing technology has been successfully utilized for building 3D structures; however, the poor mechanical properties of cell-laden hydrogels are a major concern. Here, we demonstrate the printing of a 3D cell-laden construct and its application to liver tissue engineering using 3D cell printing technology through a multi-head tissue/organ building system. Polycaprolactone (PCL) was used as a framework material because of its excellent mechanical properties. Collagen bioink containing three different types of cells-hepatocytes (HCs), human umbilical vein endothelial cells , and human lung fibroblasts--was infused into the canals of a PCL framework to induce the formation of capillary--like networks and liver cell growth. A co-cultured 3D microenvironment of the three types of cells was successfully established and maintained. The vascular formation and functional abilities of HCs (i.e., albumin secretion and urea synthesis) demonstrated that the heterotypic interaction among HCs and nonparenchymal cells increased the survivability and functionality of HCs within the collagen gel. Therefore, our results demonstrate the prospect of using cell printing technology for the creation of heterotypic cellular interaction within a structure for liver tissue engineering.


Assuntos
Órgãos Bioartificiais , Fígado/irrigação sanguínea , Fígado/crescimento & desenvolvimento , Neovascularização Fisiológica/fisiologia , Impressão Tridimensional , Engenharia Tecidual/instrumentação , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Células Cultivadas , Técnicas de Cocultura/instrumentação , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Desenho de Equipamento , Análise de Falha de Equipamento , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Hepatócitos/citologia , Hepatócitos/fisiologia , Humanos , Técnicas de Cultura de Órgãos/instrumentação , Ratos , Ratos Sprague-Dawley , Alicerces Teciduais
15.
J Biomed Mater Res A ; 104(7): 1797-804, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26991030

RESUMO

In cartilage tissue engineering, electromagnetic field (EMF) therapy has been reported to have a modest effect on promoting cartilage regeneration. However, these studies were conducted using different frequencies of EMF to stimulate chondrocytes. Thus, it is necessary to investigate the effect of EMF frequency on cartilage formation. In addition to the stimulation, a scaffold is required to satisfy the characteristics of cartilage such as its hydrated and dense extracellular matrix, and a mechanical resilience to applied loads. Therefore, we 3D-printed a composite construct composed of a polymeric framework and a chondrocyte-laden hydrogel. Here, we observed frequency-dependent positive and negative effects on chondrogenesis using a 3D cell-printed cartilage tissue. We found that a frequency of 45 Hz promoted gene expression and secretion of extracellular matrix molecules of chondrocytes. In contrast, a frequency of 7.5 Hz suppressed chondrogenic differentiation in vitro. Additionally, the EMF-treated composite constructs prior to implantation showed consistent results with those of in vitro, suggesting that in vitro pre-treatment with different EMF frequencies provides different capabilities for the enhancement of cartilage formation in vivo. This correlation between EMF frequency and 3D-printed chondrocytes suggests the necessity for optimization of EMF parameters when this physical stimulus is applied to engineered cartilage. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1797-1804, 2016.


Assuntos
Condrócitos/citologia , Campos Eletromagnéticos , Impressão Tridimensional , Alicerces Teciduais/química , Animais , Linhagem Celular , Condrogênese , Colágeno Tipo II/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
16.
Bone ; 92: 180-188, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27601226

RESUMO

Sclerostin (Sost) is a negative regulator of bone formation that acts upon the Wnt signaling pathway. Sost is mechanically regulated at both mRNA and protein level such that loading represses and unloading enhances Sost expression, in osteocytes and in circulation. The non-coding evolutionarily conserved enhancer ECR5 has been previously reported as a transcriptional regulatory element required for modulating Sost expression in osteocytes. Here we explored the mechanisms by which ECR5, or several other putative transcriptional enhancers regulate Sost expression, in response to mechanical stimulation. We found that in vivo ulna loading is equally osteoanabolic in wildtype and Sost-/- mice, although Sost is required for proper distribution of load-induced bone formation to regions of high strain. Using Luciferase reporters carrying the ECR5 non-coding enhancer and heterologous or homologous hSOST promoters, we found that ECR5 is mechanosensitive in vitro and that ECR5-driven Luciferase activity decreases in osteoblasts exposed to oscillatory fluid flow. Yet, ECR5-/- mice showed similar magnitude of load-induced bone formation and similar periosteal distribution of bone formation to high-strain regions compared to wildtype mice. Further, we found that in contrast to Sost-/- mice, which are resistant to disuse-induced bone loss, ECR5-/- mice lose bone upon unloading to a degree similar to wildtype control mice. ECR5 deletion did not abrogate positive effects of unloading on Sost, suggesting that additional transcriptional regulators and regulatory elements contribute to load-induced regulation of Sost.


Assuntos
Adaptação Fisiológica/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Glicoproteínas/deficiência , Osteócitos/fisiologia , Osteogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fenômenos Biomecânicos/fisiologia , Proteínas Morfogenéticas Ósseas/deficiência , Proteínas Morfogenéticas Ósseas/genética , Feminino , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA não Traduzido/genética
17.
J Control Release ; 238: 231-241, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-27288878

RESUMO

Since recurrence and metastasis of pancreatic cancer has a worse prognosis, chemotherapy has been typically performed to attack the remained malignant cells after resection. However, it is difficult to achieve the therapeutic concentration at the tumor site with systemic chemotherapy. Numerous local drug delivery systems have been studied to overcome the shortcomings of systemic delivery. However, because most systems involve dissolution of the drug within the carrier, the concentration of the drug is limited to the saturation solubility, and consequently cannot reach the sufficient drug dose. Therefore, we hypothesized that 3D printing of a biodegradable patch incorporated with a high drug concentration would provide a versatile shape to be administered at the exact tumor site as well as an appropriate therapeutic drug concentration with a controlled release. Here, we introduce the 3D-printed patches composed of a blend of poly(lactide-co-glycolide), polycaprolactone, and 5-fluorouracil for delivering the anti-cancer drug in a prolonged controlled manner and therapeutic dose. 3D printing technology can manipulate the geometry of the patch and the drug release kinetics. The patches were flexible, and released the drug over four weeks, and thereby suppressed growth of the subcutaneous pancreatic cancer xenografts in mice with minimized side effects. Our approach reveals that 3D printing of bioabsorbable implants containing anti-cancer drugs could be a powerful method for an effective local delivery of chemotherapeutic agents to treatment of cancers.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Preparações de Ação Retardada/química , Sistemas de Liberação de Medicamentos/instrumentação , Fluoruracila/administração & dosagem , Pâncreas/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Poliglactina 910/química , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Desenho de Equipamento , Fluoruracila/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Poliésteres/química , Impressão Tridimensional/instrumentação
18.
Arch Pharm Res ; 38(2): 272-81, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24849033

RESUMO

Transforming growth factor-ß1 (TGFß1) induces epithelial-to-mesenchymal transition (EMT) in cultured renal tubular epithelial cells. This phenotypic transition has been known to be involved in the development of chronic kidney diseases by activating profibrotic gene expression. Since oxidative stress has been recognized as one of the contributors to this TGFß1-mediated pathology, we investigated the potential involvement of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is a key transcription factor for the regulation of multiple antioxidant genes, in TGFß1-stimulated EMT gene changes using the rat proximal tubular epithelial cell line NRK52E. The treatment of NRK52E with TGFß1 led to changes in EMT gene expression, including increased α-Sma and decreased E-cadherin expression. In these cells, the TGFß1 treatment decreased the transcript level of the catalytic subunit of γ-glutamate cysteine ligase (Gclc), a glutathione (GSH) biosynthetic enzyme, and reduced the total GSH content with a concomitant decrease in Nrf2 transcription activity. Accordantly, pre-incubation with the GSH precursor N-acetylcysteine attenuated TGFß1-stimulated EMT gene changes. The involvement of Nrf2 in EMT gene changes has been demonstrated using NRK52E cells with nrf2 knockdown or pharmacological activation. When the expression of Nrf2 was stably silenced in NRK52E cells using interfering RNA administration, Gclc expression was significantly reduced and the increase in the levels of α-Sma and fibronectin-1 by TGFß1 was greater than those in the nonspecific RNA control group. Conversely, Nrf2 activation and subsequent Gclc increase by Nrf2-activating sulforaphane alleviated the TGFß1-stimulated α-Sma increase and E-cadherin decrease. Collectively, these results indicate that Nrf2-GSH signaling can modulate TGFß1-stimulated EMT gene changes and further suggest a beneficial role of Nrf2 inducers in renal pathogenesis.


Assuntos
Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glutationa/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Ratos , Proteínas Recombinantes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
19.
J Tissue Eng Regen Med ; 9(10): 1161-71, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23281198

RESUMO

An electromagnetic field is an effective stimulation tool because it promotes bone defect healing, albeit in an unknown way. Although electromagnetic fields are used for treatment after surgery, many patients prefer cell-based tissue regeneration procedures that do not require daily treatments. This study addressed the effects of an electromagnetic field on adipose-derived stem cells (ASCs) to investigate the feasibility of pretreatment to accelerate bone regeneration. After identifying a uniform electromagnetic field inside a solenoid coil, we observed that a 45 Hz electromagnetic field induced osteogenic marker expression via bone morphogenetic protein, transforming growth factor ß, and Wnt signalling pathways based on microarray analyses. This electromagnetic field increased osteogenic gene expression, alkaline phosphate activity and nodule formation in vitro within 2 weeks, indicating that this pretreatment may provide osteogenic potential to ASCs on three-dimensional (3D) ceramic scaffolds. This pretreatment effect of an electromagnetic field resulted in significantly better bone regeneration in a mouse calvarial defect model over 4 weeks compared to that in the untreated group. This short-term evaluation showed that the electromagnetic field pretreatment may be a future therapeutic option for bone defect treatment.


Assuntos
Tecido Adiposo/citologia , Campos Eletromagnéticos , Consolidação da Fratura , Células-Tronco/citologia , Animais , Transplante de Células , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais
20.
Artigo em Inglês | MEDLINE | ID: mdl-25653639

RESUMO

Mechanical loading is essential to maintain normal bone metabolism and the balance between bone formation and resorption. The cellular mechanisms that control mechanotransduction are not fully defined, but several key pathways have been identified. We discuss the roles of several components of the Wnt signaling cascade, namely Lrp5, Lrp6, and ß-catenin in mechanical loading-induced bone formation. Lrp5 is an important Wnt co-receptor for regulating bone mass and mechanotransduction, and appears to function principally by augmenting bone formation. Lrp6 also regulates bone mass but its action might involve resorption as well as formation. The role of Lrp6 in mechanotransduction is unclear. Studies addressing the role of ß-catenin in bone metabolism and mechanotransduction highlight the uncertainties in downstream modulators of Lrp5 and Lrp6. Taken together, these data indicate that mechanical loading might affect bone regulation triggering the canonical Wnt signaling (and perhaps other pathways) not only via Lrp5 but also via Lrp6. Further work is needed to clarify the role of the Wnt signaling pathway in Lrp5 and/or Lrp6-mediated mechanotransduction, which could eventually lead to powerful therapeutic agents that might mimic the anabolic effects of mechanical stimulation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA