Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Microgravity ; 9(1): 75, 2023 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-37723136

RESUMO

Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).

2.
Cell Rep ; 42(5): 112299, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37080202

RESUMO

Understanding the axis of the human microbiome and physiological homeostasis is an essential task in managing deep-space-travel-associated health risks. The NASA-led Rodent Research 5 mission enabled an ancillary investigation of the gut microbiome, varying exposure to microgravity (flight) relative to ground controls in the context of previously shown bone mineral density (BMD) loss that was observed in these flight groups. We demonstrate elevated abundance of Lactobacillus murinus and Dorea sp. during microgravity exposure relative to ground control through whole-genome sequencing and 16S rRNA analyses. Specific functionally assigned gene clusters of L. murinus and Dorea sp. capable of producing metabolites, lactic acid, leucine/isoleucine, and glutathione are enriched. These metabolites are elevated in the microgravity-exposed host serum as shown by liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. Along with BMD loss, ELISA reveals increases in osteocalcin and reductions in tartrate-resistant acid phosphatase 5b signifying additional loss of bone homeostasis in flight.


Assuntos
Microbioma Gastrointestinal , Voo Espacial , Humanos , RNA Ribossômico 16S/genética , Cromatografia Líquida , Viagem , Espectrometria de Massas em Tandem
3.
Biomaterials ; 287: 121609, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35839586

RESUMO

Recent investigations into mechanisms behind the development of osteoporosis suggest that suppressing PPARγ-mediated adipogenesis can improve bone formation and bone mineral density. In this study, we investigated a co-treatment strategy to enhance bone formation by combining NELL-1, an osteogenic molecule that has been extensively studied for its potential use as a therapeutic for osteoporosis, with two methods of PPARγ suppression. First, we suppressed PPARγ genetically using lentiviral PPARγ-shRNA in immunocompromised mice for a proof of concept. Second, we used a PPARγ antagonist to suppress PPARγ pharmacologically in immunocompetent senile osteopenic mice for clinical transability. We found that the co-treatment strategy significantly increased bone formation, increased the proliferation stage cell population, decreased late apoptosis of primary mouse BMSCs, and increased osteogenic marker mRNA levels in comparison to the single agent treatment groups. The addition of PPARγ suppression to NELL-1 therapy enhanced NELL-1's effects on bone formation by upregulating anabolic processes without altering NELL-1's inhibitory effects on osteoclastic and adipogenic activities. Our findings suggest that combining PPARγ suppression with therapeutic NELL-1 may be a viable method that can be further developed as a novel strategy to reverse bone loss and decrease marrow adiposity in age-related osteoporosis.

4.
Methods Mol Biol ; 2235: 127-137, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33576974

RESUMO

Human pericytes are a perivascular cell population with mesenchymal stem cell properties, present in all vascularized tissues. Human pericytes have a distinct immunoprofile, which may be leveraged for purposes of cell purification. Adipose tissue is the most commonly used cell source for human pericyte derivation. Pericytes can be isolated by FACS (fluorescence-activated cell sorting), most commonly procured from liposuction aspirates. Pericytes have clonal multilineage differentiation potential, and their potential utility for bone regeneration has been described across multiple animal models. The following review will discuss in vivo methods for assessing the bone-forming potential of purified pericytes. Potential models include (1) mouse intramuscular implantation, (2) mouse calvarial defect implantation, and (3) rat spinal fusion models. In addition, the presented surgical protocols may be used for the in vivo analysis of other osteoprogenitor cell types.


Assuntos
Células da Medula Óssea/metabolismo , Pericitos/metabolismo , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Regeneração Óssea/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Separação Celular/métodos , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Osteogênese/fisiologia , Pericitos/citologia , Ratos
5.
J Bone Miner Res ; 34(3): 533-546, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30352124

RESUMO

NELL-1, an osteoinductive protein, has been shown to regulate skeletal ossification. Interestingly, an interstitial 11p14.1-p15.3 deletion involving the Nell-1 gene was recently reported in a patient with short stature and delayed fontanelle closure. Here we sought to define the role of Nell-1 in endochondral ossification by investigating Nell-1-specific inactivation in Col2α1-expressing cell lineages. Nell-1flox/flox ; Col2α1-Cre+ (Nell-1Col2α1 KO) mice were generated for comprehensive analysis. Nell-1Col2α1 KO mice were born alive but displayed subtle femoral length shortening. At 1 and 3 months postpartum, Nell-1 inactivation resulted in dwarfism and premature osteoporotic phenotypes. Specifically, Nell-1Col2α1 KO femurs and tibias exhibited significantly reduced length, bone mineral density (BMD), bone volume per tissue volume (BV/TV), trabecular number/thickness, cortical volume/thickness/density, and increased trabecular separation. The decreased bone formation rate revealed by dynamic histomorphometry was associated with altered numbers and/or function of osteoblasts and osteoclasts. Furthermore, longitudinal observations by in vivo micro-CT showed delayed and reduced mineralization at secondary ossification centers in mutants. Histologically, reduced staining intensities of Safranin O, Col-2, Col-10, and fewer BrdU-positive chondrocytes were observed in thinner Nell-1Col2α1 KO epiphyseal plates along with altered distribution and weaker expression level of Ihh, Patched-1, PTHrP, and PTHrP receptor. Primary Nell-1Col2α1 KO chondrocytes also exhibited decreased proliferation and differentiation, and its downregulated expression of the Ihh-PTHrP signaling molecules can be partially rescued by exogenous Nell-1 protein. Moreover, intranuclear Gli-1 protein and gene expression of the Gli-1 downstream target genes, Hip-1 and N-Myc, were also significantly decreased with Nell-1 inactivation. Notably, the rescue effects were diminished/reduced with application of Ihh signaling inhibitors, cyclopamine or GANT61. Taken together, these findings suggest that Nell-1 is a pivotal modulator of epiphyseal homeostasis and endochondral ossification. The cumulative chondrocyte-specific Nell-1 inactivation significantly impedes appendicular skeletogenesis resulting in dwarfism and premature osteoporosis through inhibiting Ihh signaling and predominantly altering the Ihh-PTHrP feedback loop. © 2018 American Society for Bone and Mineral Research.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Condrócitos/metabolismo , Nanismo/metabolismo , Osteogênese , Osteoporose/metabolismo , Animais , Condrócitos/patologia , Nanismo/diagnóstico por imagem , Nanismo/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Knockout , Osteoporose/diagnóstico por imagem , Osteoporose/genética , Osteoporose/patologia , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Microtomografia por Raio-X
6.
Am J Pathol ; 189(3): 648-664, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30593824

RESUMO

The Food and Drug Administration-approved clinical dose (1.5 mg/mL) of bone morphogenetic protein-2 (BMP2) has been reported to induce significant adverse effects, including cyst-like adipose-infiltrated abnormal bone formation. These undesirable complications occur because of increased adipogenesis, at the expense of osteogenesis, through BMP2-mediated increases in the master regulatory gene for adipogenesis, peroxisome proliferator-activated receptor-γ (PPARγ). Inhibiting PPARγ during osteogenesis has been suggested to drive the differentiation of bone marrow stromal/stem cells toward an osteogenic, rather than an adipogenic, lineage. We demonstrate that knocking down PPARγ while concurrently administering BMP2 can reduce adipogenesis, but we found that it also impairs BMP2-induced osteogenesis and leads to bone nonunion in a mouse femoral segmental defect model. In addition, in vitro studies using the mouse bone marrow stromal cell line M2-10B4 and mouse primary bone marrow stromal cells confirmed that PPARγ knockdown inhibits BMP2-induced adipogenesis; attenuates BMP2-induced cell proliferation, migration, invasion, and osteogenesis; and escalates BMP2-induced cell apoptosis. More important, BMP receptor 2 and 1B expression was also significantly inhibited by the combined BMP2 and PPARγ knockdown treatment. These findings indicate that PPARγ is critical for BMP2-mediated osteogenesis during bone repair. Thus, uncoupling BMP2-mediated osteogenesis and adipogenesis using PPARγ inhibition to combat BMP2's adverse effects may not be feasible.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea , Fêmur , Osteogênese , PPAR gama/metabolismo , Adipogenia/genética , Animais , Proteína Morfogenética Óssea 2/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Fêmur/lesões , Fêmur/metabolismo , Fêmur/patologia , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Transgênicos , PPAR gama/genética
7.
Prog Orthod ; 19(1): 41, 2018 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-30345476

RESUMO

BACKGROUND: Miniscrew-assisted rapid palatal expansion (MARPE) has been adopted in recent years to expand the maxilla in late adolescence and adult patients. Maxillary Skeletal Expander (MSE) is a device that exploits the principles of skeletal anchorage to transmit the expansion force directly to the maxillary bony structures and is characterized by the miniscrews' engagement of the palatal and nasal cortical bone layers. In the literature, it has been reported that the zygomatic buttress is a major constraint that hampers the lateral movement of maxilla, since maxilla is located medially to the zygomatic arches. The objective of the present study is to analyze the changes in the zygomatic bone, maxillary bone, and zygomatic arches and to localize the center of rotation for the zygomaticomaxillary complex in the horizontal plane after treatment with MSE, using high-resolution cone-beam computed tomography (CBCT) images. METHODS: Fifteen subjects with a mean age of 17.2 (± 4.2) years were treated with MSE. CBCT records were taken before and after miniscrew-assisted maxillary expansion; three linear and four angular parameters were identified in the axial zygomatic section (AZS) and were compared from pre-treatment to post-treatment using the Wilcoxon signed rank test. RESULTS: Anterior inter-maxillary distance increased by 2.8 mm, posterior inter-zygomatic distance by 2.4 mm, angle of the zygomatic process of the temporal bone by 1.7° and 2.1° (right and left side) (P < 0.01). Changes in posterior inter-temporal distance and zygomaticotemporal angle were negligible (P > 0.05). CONCLUSIONS: In the horizontal plane, the maxillary and zygomatic bones and the whole zygomatic arch were significantly displaced in a lateral direction after treatment with MSE. The center of rotation for the zygomaticomaxillary complex was located near the proximal portion of the zygomatic process of the temporal bone, more posteriorly and more laterally than what has been reported in the literature for tooth-borne expanders. Bone bending takes place in the zygomatic process of the temporal bone during miniscrew-supported maxillary expansion.


Assuntos
Parafusos Ósseos , Tomografia Computadorizada de Feixe Cônico , Maxila/diagnóstico por imagem , Maxila/fisiologia , Técnica de Expansão Palatina/instrumentação , Zigoma/diagnóstico por imagem , Zigoma/fisiologia , Adolescente , Feminino , Humanos , Imageamento Tridimensional , Masculino , Má Oclusão/terapia , Mandíbula/diagnóstico por imagem , Desenho de Aparelho Ortodôntico , Estudos Retrospectivos , Rotação , Adulto Jovem
8.
Am J Orthod Dentofacial Orthop ; 154(3): 337-345, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30173836

RESUMO

INTRODUCTION: Our objectives were to evaluate midfacial skeletal changes in the coronal plane and the implications of circummaxillary sutures and to localize the center of rotation for the zygomaticomaxillary complex after therapy with a bone-anchored maxillary expander, using high-resolution cone-beam computed tomography. METHODS: Fifteen subjects with a mean age of 17.2 ± 4.2 years were treated with a bone-anchored maxillary expander. Pretreatment and posttreatment cone-beam computed tomography images were superimposed and examined for comparison. RESULTS: Upper interzygomatic distance increased by 0.5 mm, lower interzygomatic distance increased by 4.6 mm, frontozygomatic angles increased by 2.5° and 2.9° (right and left sides), maxillary inclinations increased by 2.0° and 2.5° (right and left sides), and intermolar distance increased by 8.3 mm (P <0.05). Changes in frontoethmoidal, zygomaticomaxillary, and molar basal bone angles were negligible (P >0.05). CONCLUSIONS: A significant lateral displacement of the zygomaticomaxillary complex occurred in late adolescent patients treated with a bone-anchored maxillary expander. The zygomatic bone tended to rotate outward along with the maxilla with a common center of rotation located near the superior aspect of the frontozygomatic suture. Dental tipping of the molars was negligible during treatment.


Assuntos
Tomografia Computadorizada de Feixe Cônico , Suturas Cranianas/diagnóstico por imagem , Suturas Cranianas/fisiologia , Ossos Faciais/diagnóstico por imagem , Ossos Faciais/fisiologia , Técnica de Expansão Palatina , Âncoras de Sutura , Adolescente , Adulto , Feminino , Humanos , Imageamento Tridimensional , Masculino , Estudos Retrospectivos
9.
Artigo em Inglês | MEDLINE | ID: mdl-29201497

RESUMO

Blocking transforming growth factor (TGF)ß1 signal transduction has been a central strategy for scar reduction; however, this approach appears to be minimally effective. Here, we show that fibromodulin (FMOD), a 59-kD small leucine-rich proteoglycan critical for normal collagen fibrillogenesis, significantly reduces scar formation while simultaneously increasing scar strength in both adult rodent models and porcine wounds, which simulate human cutaneous scar repair. Mechanistically, FMOD uncouples pro-migration/contraction cellular signals from pro-fibrotic signaling by selectively enhancing SMAD3-mediated signal transduction, while reducing AP-1-mediated TGFß1 auto-induction and fibrotic extracellular matrix accumulation. Consequently, FMOD accelerates TGFß1-responsive adult fibroblast migration, myofibroblast conversion, and function. Furthermore, our findings strongly indicate that, by delicately orchestrating TGFß1 activities rather than indiscriminately blocking TGFß1, FMOD elicits fetal-like cellular and molecular phenotypes in adult dermal fibroblasts in vitro and adult cutaneous wounds in vivo, which is a unique response of living system undescribed previously. Taken together, this study illuminates the signal modulating activities of FMOD beyond its structural support functions, and highlights the potential for FMOD-based therapies to be used in cutaneous wound repair.

10.
Prog Orthod ; 18(1): 34, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29090368

RESUMO

BACKGROUND: Mini-implant-assisted rapid palatal expansion (MARPE) appliances have been developed with the aim to enhance the orthopedic effect induced by rapid maxillary expansion (RME). Maxillary Skeletal Expander (MSE) is a particular type of MARPE appliance characterized by the presence of four mini-implants positioned in the posterior part of the palate with bi-cortical engagement. The aim of the present study is to evaluate the MSE effects on the midpalatal and pterygopalatine sutures in late adolescents, using high-resolution CBCT. Specific aims are to define the magnitude and sagittal parallelism of midpalatal suture opening, to measure the extent of transverse asymmetry of split, and to illustrate the possibility of splitting the pterygopalatine suture. METHODS: Fifteen subjects (mean age of 17.2 years; range, 13.9-26.2 years) were treated with MSE. Pre- and post-treatment CBCT exams were taken and superimposed. A novel methodology based on three new reference planes was utilized to analyze the sutural changes. Parameters were compared from pre- to post-treatment and between genders non-parametrically using the Wilcoxon sign rank test. For the frequency of openings in the lower part of the pterygopalatine suture, the Fisher's exact test was used. RESULTS: Regarding the magnitude of midpalatal suture opening, the split at anterior nasal spine (ANS) and at posterior nasal spine (PNS) was 4.8 and 4.3 mm, respectively. The amount of split at PNS was 90% of that at ANS, showing that the opening of the midpalatal suture was almost perfectly parallel antero-posteriorly. On average, one half of the anterior nasal spine (ANS) moved more than the contralateral one by 1.1 mm. Openings between the lateral and medial plates of the pterygoid process were detectable in 53% of the sutures (P < 0.05). No significant differences were found in the magnitude and frequency of suture opening between males and females. Correlation between age and suture opening was negligible (R 2 range, 0.3-4.2%). CONCLUSIONS: Midpalatal suture was successfully split by MSE in late adolescents, and the opening was almost perfectly parallel in a sagittal direction. Regarding the extent of transverse asymmetry of the split, on average one half of ANS moved more than the contralateral one by 1.1 mm. Pterygopalatine suture was split in its lower region by MSE, as the pyramidal process was pulled out from the pterygoid process. Patient gender and age had a negligible influence on suture opening for the age group considered in the study.


Assuntos
Tomografia Computadorizada de Feixe Cônico/métodos , Imageamento Tridimensional/métodos , Técnica de Expansão Palatina/instrumentação , Palato/diagnóstico por imagem , Fossa Pterigopalatina/diagnóstico por imagem , Adolescente , Adulto , Feminino , Humanos , Masculino , Mandíbula/anatomia & histologia , Mandíbula/diagnóstico por imagem , Maxila/anatomia & histologia , Maxila/diagnóstico por imagem , Desenho de Aparelho Ortodôntico , Palato/anatomia & histologia , Fossa Pterigopalatina/anatomia & histologia , Estudos Retrospectivos , Adulto Jovem
11.
Proc Natl Acad Sci U S A ; 114(35): E7218-E7225, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808036

RESUMO

Patients with cleft lip and/or palate (CLP), who undergo numerous medical interventions from infancy, can suffer from lifelong debilitation caused by underdeveloped maxillae. Conventional treatment approaches use maxillary expansion techniques to develop normal speech, achieve functional occlusion for nutrition intake, and improve esthetics. However, as patients with CLP congenitally lack bone in the cleft site with diminished capacity for bone formation in the expanded palate, more than 80% of the patient population experiences significant postexpansion relapse. While such relapse has been a long-standing battle in craniofacial care of patients, currently there are no available strategies to address this pervasive problem. Estrogen, 17ß-estradiol (E2), is a powerful therapeutic agent that plays a critical role in bone homeostasis. However, E2's clinical application is less appreciated due to several limitations, including its pleiotropic effects and short half-life. Here, we developed a treatment strategy using an injectable system with photo-cross-linkable hydrogel (G) and nanodiamond (ND) technology to facilitate the targeted and sustained delivery of E2 to promote bone formation. In a preclinical expansion/relapse model, this functionalized E2/ND/G complex substantially reduced postexpansion relapse by nearly threefold through enhancements in sutural remodeling compared with unmodified E2 administration. The E2/ND/G group demonstrated greater bone volume by twofold and higher osteoblast number by threefold, compared with the control group. The E2/ND/G platform maximized the beneficial effects of E2 through its extended release with superior efficacy and safety at the local level. This broadly applicable E2 delivery platform shows promise as an adjuvant therapy in craniofacial care of patients.


Assuntos
Estrogênios/farmacologia , Nanodiamantes/uso terapêutico , Técnica de Expansão Palatina/instrumentação , Animais , Fenda Labial/cirurgia , Fissura Palatina/terapia , Modelos Animais de Doenças , Feminino , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Nanoestruturas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Recidiva , Prevenção Secundária/métodos , Resultado do Tratamento
12.
JCI Insight ; 2(12)2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28614787

RESUMO

NELL-1 is a secreted, osteogenic protein first discovered to control ossification of the cranial skeleton. Recently, NELL-1 has been implicated in bone maintenance. However, the cellular determinants of NELL-1's bone-forming effects are still unknown. Here, recombinant human NELL-1 (rhNELL-1) implantation was examined in a clinically relevant nonhuman primate lumbar spinal fusion model. Prolonged rhNELL-1 protein release was achieved using an apatite-coated ß-tricalcium phosphate carrier, resulting in a local influx of stem cell antigen-1-positive (Sca-1+) mesenchymal progenitor cells (MPCs), and complete osseous fusion across all samples (100% spinal fusion rate). Murine studies revealed that Nell-1 haploinsufficiency results in marked reductions in the numbers of Sca-1+CD45-CD31- bone marrow MPCs associated with low bone mass. Conversely, rhNELL-1 systemic administration in mice showed a marked anabolic effect accompanied by increased numbers of Sca-1+CD45-CD31- bone marrow MPCs. Mechanistically, rhNELL-1 induces Sca-1 transcription among MPCs, in a process requiring intact Wnt/ß-catenin signaling. In summary, NELL-1 effectively induces bone formation across small and large animal models either via local implantation or intravenous delivery. NELL-1 induces an expansion of a bone marrow subset of MPCs with Sca-1 expression. These findings provide compelling justification for the clinical translation of a NELL-1-based therapy for local or systemic bone formation.

13.
Am J Pathol ; 187(7): 1485-1495, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28502475

RESUMO

Multiple case reports using recombinant human bone morphogenetic protein-2 (rhBMP-2) have reported complications. However, the local adverse effects of rhBMP-2 application are not well documented. In this report we show that, in addition to promoting lumbar spinal fusion through potent osteogenic effects, rhBMP-2 augmentation promotes local cyst-like osteolytic formations in sheep trabecular bones that have undergone anterior lumbar interbody fusion. Three months after operation, conventional computed tomography showed that the trabecular bones of the rhBMP-2 application groups could fuse, whereas no fusion was observed in the control group. Micro-computed tomography analysis revealed that the core implant area's bone volume fraction and bone mineral density increased proportionately with rhBMP-2 dose. Multiple cyst-like bone voids were observed in peri-implant areas when using rhBMP-2 applications, and these sites showed significant bone mineral density decreases in relation to the unaffected regions. Biomechanically, these areas decreased in strength by 32% in comparison with noncystic areas. Histologically, rhBMP-2-affected void sites had an increased amount of fatty marrow, thinner trabecular bones, and significantly more adiponectin- and cathepsin K-positive cells. Despite promoting successful fusion, rhBMP-2 use in clinical applications may result in local adverse structural alterations and compromised biomechanical changes to the bone.


Assuntos
Proteína Morfogenética Óssea 2/administração & dosagem , Vértebras Lombares/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fusão Vertebral/métodos , Fator de Crescimento Transformador beta/administração & dosagem , Animais , Densidade Óssea/efeitos dos fármacos , Proteína Morfogenética Óssea 2/efeitos adversos , Proteína Morfogenética Óssea 2/genética , Feminino , Humanos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia , Modelos Animais , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/genética , Ovinos , Fusão Vertebral/efeitos adversos , Tomografia Computadorizada por Raios X , Fator de Crescimento Transformador beta/efeitos adversos , Fator de Crescimento Transformador beta/genética
14.
Plast Reconstr Surg ; 139(6): 1385-1396, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28198775

RESUMO

BACKGROUND: Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS: A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS: Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS: In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/administração & dosagem , Fraturas Ósseas/terapia , Glicoproteínas/administração & dosagem , Proteínas Hedgehog/administração & dosagem , Osteogênese/efeitos dos fármacos , Animais , Biópsia por Agulha , Terapia Combinada , Modelos Animais de Doenças , Feminino , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Distribuição Aleatória , Estatísticas não Paramétricas , Osso Temporal/cirurgia , Alicerces Teciduais
15.
Tissue Eng Part A ; 22(23-24): 1357-1366, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27702396

RESUMO

Hedgehog (Hh) signaling positively regulates both endochondral and intramembranous ossification. Use of small molecules for tissue engineering applications poses several advantages. In this study, we examined whether use of an acellular scaffold treated with the small molecule Smoothened agonist (SAG) could aid in critical-size mouse calvarial defect repair. First, we verified the pro-osteogenic effect of SAG in vitro, using primary neonatal mouse calvarial cells (NMCCs). Next, a 4 mm nonhealing defect was created in the mid-parietal bone of 10-week-old CD-1 mice. The scaffold consisted of a custom-fabricated poly(lactic-co-glycolic acid) disc with hydroxyapatite coating (measuring 4 mm diameter × 0.5 mm thickness). Treatment groups included dimethylsulfoxide control (n = 6), 0.5 mM SAG (n = 7) or 1.0 mM SAG (n = 7). Evaluation was performed at 4 and 8 weeks postoperative, by a combination of high-resolution microcomputed tomography, histology (H & E, Masson's Trichrome), histomorphometry, and immunohistochemistry (BSP, OCN, VEGF). In vivo results showed that SAG treatment induced a significant and dose-dependent increase in calvarial bone healing by all radiographic parameters. Histomorphometric analysis showed an increase in all parameters of bone formation with SAG treatment, but also an increase in blood vessel number and density. In summary, SAG is a pro-osteogenic, provasculogenic stimulus when applied locally in a bone defect environment.


Assuntos
Cicloexilaminas/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Consolidação da Fratura/efeitos dos fármacos , Osso Parietal/lesões , Osso Parietal/metabolismo , Tiofenos/farmacologia , Alicerces Teciduais/química , Animais , Masculino , Camundongos , Osso Parietal/diagnóstico por imagem , Osso Parietal/patologia
16.
Tissue Eng Part C Methods ; 22(5): 451-63, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26956416

RESUMO

Trabecular bone is frequently studied in osteoporosis research because changes in trabecular bone are the most common cause of osteoporotic fractures. Dual energy X-ray absorptiometry (DXA) analysis specific to trabecular bone-rich regions is crucial to longitudinal osteoporosis research. The purpose of this study is to define a novel method for accurately analyzing trabecular bone-rich regions in mice via DXA. This method will be utilized to analyze scans obtained from the International Space Station in an upcoming study of microgravity-induced bone loss. Thirty 12-week-old BALB/c mice were studied. The novel method was developed by preanalyzing trabecular bone-rich sites in the distal femur, proximal tibia, and lumbar vertebrae via high-resolution X-ray imaging followed by DXA and micro-computed tomography (micro-CT) analyses. The key DXA steps described by the novel method were (1) proper mouse positioning, (2) region of interest (ROI) sizing, and (3) ROI positioning. The precision of the new method was assessed by reliability tests and a 14-week longitudinal study. The bone mineral content (BMC) data from DXA was then compared to the BMC data from micro-CT to assess accuracy. Bone mineral density (BMD) intra-class correlation coefficients of the new method ranging from 0.743 to 0.945 and Levene's test showing that there was significantly lower variances of data generated by new method both verified its consistency. By new method, a Bland-Altman plot displayed good agreement between DXA BMC and micro-CT BMC for all sites and they were strongly correlated at the distal femur and proximal tibia (r=0.846, p<0.01; r=0.879, p<0.01, respectively). The results suggest that the novel method for site-specific analysis of trabecular bone-rich regions in mice via DXA yields more precise, accurate, and repeatable BMD measurements than the conventional method.


Assuntos
Absorciometria de Fóton/métodos , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Microtomografia por Raio-X/métodos , Animais , Densidade Óssea , Feminino , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos BALB C , Osteoporose/prevenção & controle
17.
Biomaterials ; 83: 194-206, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26774565

RESUMO

Pluripotent or multipotent cell-based therapeutics are vital for skeletal reconstruction in non-healing critical-sized defects since the local endogenous progenitor cells are not often adequate to restore tissue continuity or function. However, currently available cell-based regenerative strategies are hindered by numerous obstacles including inadequate cell availability, painful and invasive cell-harvesting procedures, and tumorigenesis. Previously, we established a novel platform technology for inducing a quiescent stem cell-like stage using only a single extracellular proteoglycan, fibromodulin (FMOD), circumventing gene transduction. In this study, we further purified and significantly increased the reprogramming rate of the yield multipotent FMOD reprogrammed (FReP) cells. We also exposed the 'molecular blueprint' of FReP cell osteogenic differentiation by gene profiling. Radiographic analysis showed that implantation of FReP cells into a critical-sized SCID mouse calvarial defect, contributed to the robust osteogenic capability of FReP cells in a challenging clinically relevant traumatic scenario in vivo. The persistence, engraftment, and osteogenesis of transplanted FReP cells without tumorigenesis in vivo were confirmed by histological and immunohistochemical staining. Taken together, we have provided an extended potency, safety, and molecular profile of FReP cell-based bone regeneration. Therefore, FReP cells present a high potential for cellular and gene therapy products for bone regeneration.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/farmacologia , Proteoglicanas/farmacologia , Animais , Células CHO , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Transplante de Células , Células Cultivadas , Cricetinae , Cricetulus , Meios de Cultura/farmacologia , Fibromodulina , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Camundongos SCID , Minerais/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Crânio/diagnóstico por imagem , Crânio/efeitos dos fármacos , Crânio/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...