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










Base de dados
Intervalo de ano de publicação
1.
J Cell Mol Med ; 22(4): 2510-2513, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29392829

RESUMO

Hypertrophic scarring is a major postoperative complication which leads to severe disfigurement and dysfunction in patients and usually requires multiple surgical revisions due to its high recurrence rates. Excessive-mechanical-loading across wounds is an important initiator of hypertrophic scarring formation. In this study, we demonstrate that intradermal administration of a single extracellular matrix (ECM) molecule-fibromodulin (FMOD) protein-can significantly reduce scar size, increase tensile strength, and improve dermal collagen architecture organization in the normal and even excessive-mechanical-loading red Duroc pig wound models. Since pig skin is recognized by the Food and Drug Administration as the closest animal equivalent to human skin, and because red Duroc pigs show scarring that closely resembles human proliferative scarring and hypertrophic scarring, FMOD-based technologies hold high translational potential and applicability to human patients suffering from scarring-especially hypertrophic scarring.


Assuntos
Cicatriz/tratamento farmacológico , Fibromodulina/administração & dosagem , Dermatopatias/tratamento farmacológico , Cicatrização/efeitos dos fármacos , Animais , Cicatriz/genética , Cicatriz/patologia , Proteínas da Matriz Extracelular/administração & dosagem , Proteínas da Matriz Extracelular/genética , Fibromodulina/genética , Humanos , Injeções Intradérmicas , Pele/efeitos dos fármacos , Pele/lesões , Dermatopatias/genética , Dermatopatias/patologia , Estresse Mecânico , Suínos , Resistência à Tração/efeitos dos fármacos , Cicatrização/genética
2.
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
3.
Am J Pathol ; 186(2): 419-34, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26772960

RESUMO

The differentiation factor NEL-like molecule-1 (NELL-1) has been reported as osteoinductive in multiple in vivo preclinical models. Bone morphogenetic protein (BMP)-2 is used clinically for skeletal repair, but in vivo administration can induce abnormal, adipose-filled, poor-quality bone. We demonstrate that NELL-1 combined with BMP2 significantly optimizes osteogenesis in a rodent femoral segmental defect model by minimizing the formation of BMP2-induced adipose-filled cystlike bone. In vitro studies using the mouse bone marrow stromal cell line M2-10B4 and human primary bone marrow stromal cells have confirmed that NELL-1 enhances BMP2-induced osteogenesis and inhibits BMP2-induced adipogenesis. Importantly, the ability of NELL-1 to direct BMP2-treated cells toward osteogenesis and away from adipogenesis requires intact canonical Wnt signaling. Overall, these studies establish the feasibility of combining NELL-1 with BMP2 to improve clinical bone regeneration and provide mechanistic insight into canonical Wnt pathway activity during NELL-1 and BMP2 osteogenesis. The novel abilities of NELL-1 to stimulate Wnt signaling and to repress adipogenesis may highlight new treatment approaches for bone loss in osteoporosis.


Assuntos
Adipogenia , Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Osteogênese/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos Endogâmicos Lew , Transdução de Sinais/fisiologia
4.
Tissue Eng Part A ; 19(21-22): 2390-401, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23758588

RESUMO

Bone-morphogenetic protein 2 (BMP2) is currently the only Food and Drug Administration-approved osteoinductive growth factor used in clinical settings for bone regeneration and repair. However, the use of BMP2 is encumbered by numerous clinical complications, including postoperative inflammation and life-threatening cervical swelling. Thus, methods to prevent BMP2-induced inflammation would have far-reaching clinical implications toward improving current BMP2-based methods for bone regeneration. For the first time, we investigate the potential role of the growth factor Nel-like molecule-1 (NELL-1) in inhibiting BMP2-induced inflammation. Adult rats underwent a femoral bone onlay procedure, treated with either BMP2 protein (4 mg/mL), NELL-1 protein (4 mg/mL), or both proteins combined. Animals were evaluated at 3, 7, and 14 days postoperatively by histology, histomorphometry, immunohistochemistry, and real-time PCR for markers of inflammation (TNFα, IL6). The relative levels of TNFα and IL6 in serum were also detected by ELISA. The mechanism for NELL-1's anti-inflammatory effect was further assessed through examining inflammatory markers and generation of reactive oxygen species (ROS) in the mouse embryonic fibroblast NIH3T3 cells. BMP2 significantly induced local inflammation, including an early and pronounced polymorphonuclear cell infiltration accompanied by increased expression of TNFα and IL6. Treatment with NELL-1 alone elicited no significant inflammatory response. However, NELL-1 significantly attenuated BMP2-induced inflammation by all markers and at all timepoints. These local findings were also confirmed using systemic serum inflammatory biomarkers (TNFα, IL6). In each case, NELL-1 fully reversed BMP2-induced systemic inflammation. Lastly, our findings were recapitulated in vitro, where NELL-1 suppressed BMP2 induced expression of inflammatory markers, as well as NF-κB transcriptional activity and generation of ROS. BMP2-induced inflammation is a serious public health concern with potentially life-threatening complications. In the present study, we observed that the growth factor, NELL-1, significantly attenuates or completely reverses BMP2-induced inflammation. The mechanisms of NELL-1's anti-inflammatory effect are only partially elucidated, and may include reduction of NF-κB transcriptional activity or ROS generation.


Assuntos
Proteína Morfogenética Óssea 2/efeitos adversos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Proteínas do Tecido Nervoso/uso terapêutico , Animais , Proteínas de Ligação ao Cálcio , Humanos , Inflamação/sangue , Interleucina-6/sangue , Masculino , Camundongos , Células NIH 3T3 , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/sangue
5.
Proc Natl Acad Sci U S A ; 110(23): 9469-74, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23690607

RESUMO

Mesenchymal stem cell (MSC)-based transplantation is a promising therapeutic approach for bone regeneration and repair. In the realm of therapeutic bone regeneration, the defect or injured tissues are frequently inflamed with an abnormal expression of inflammatory mediators. Growing evidence suggests that proinflammatory cytokines inhibit osteogenic differentiation and bone formation. Thus, for successful MSC-mediated repair, it is important to overcome the inflammation-mediated inhibition of tissue regeneration. In this study, using genetic and chemical approaches, we found that proinflammatory cytokines TNF and IL-17 stimulated IκB kinase (IKK)-NF-κB and impaired osteogenic differentiation of MSCs. In contrast, the inhibition of IKK-NF-κB significantly enhanced MSC-mediated bone formation. Mechanistically, we found that IKK-NF-κB activation promoted ß-catenin ubiquitination and degradation through induction of Smurf1 and Smurf2. To translate our basic findings to potential clinic applications, we showed that the IKK small molecule inhibitor, IKKVI, enhanced osteogenic differentiation of MSCs. More importantly, the delivery of IKKVI promoted MSC-mediated craniofacial bone regeneration and repair in vivo. Considering the well established role of NF-κB in inflammation and infection, our results suggest that targeting IKK-NF-κB may have dual benefits in enhancing bone regeneration and repair and inhibiting inflammation, and this concept may also have applicability in many other tissue regeneration situations.


Assuntos
Diferenciação Celular/fisiologia , Quinase I-kappa B/metabolismo , Células-Tronco Mesenquimais/citologia , NF-kappa B/metabolismo , Osteogênese/fisiologia , beta Catenina/metabolismo , Animais , Antraquinonas , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Humanos , Interleucina-17/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Osteogênese/efeitos dos fármacos , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos
6.
J Orthop Sci ; 18(4): 646-57, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23686083

RESUMO

BACKGROUND: Spinal fusion is among the most commonly performed orthopaedic procedures. Unfortunately, current treatments such as autologous bone grafting or recombinant proteins (BMP-2) have numerous clinical shortcomings. Here, we directly compare the efficacy of NELL-1, a novel osteoinductive growth factor, to two currently available treatments, (1) recombinant BMP-2 and (2) iliac crest bone grafting, in a spinal fusion model. METHODS: Twenty-six skeletally mature athymic rats underwent posterolateral spine fusion of L4/L5 vertebrae. Treatment groups included NELL-1 (10 and 50 µg) in a demineralized bone matrix (DBX), as compared to BMP-2 (90 µg) in an absorbable collagen sponge (ACS) or morselized iliac crest bone. Scaffolds without recombinant protein were used as controls. Animals were sacrificed at 4 weeks post-operative and fusion was assessed by manual palpation, radiography [high-resolution X-ray, micro-computed tomography (microCT)], histology (hematoxylin and eosin, Masson's trichrome) and immunohistochemistry (osteocalcin). RESULTS: Results showed 100 % fusion in all NELL-1- and BMP-2-treated samples. In contrast, lower rates of fusion were observed in scaffold-only and bone graft treatment groups. MicroCT scans revealed radiographic evidence of fusion among spines treated with NELL-1. Bone bridging was also observed with BMP-2 treatment, but was accompanied by inner radiolucency, suggesting cyst-like bone formation. Histologically, NELL-1-treated grafts showed increased bone formation, endochondral ossification and vascularization. Although BMP-2 treated grafts exhibited increased bone formation and angiogenesis, numerous adipocytes were also observed. CONCLUSION: NELL-1-based bone grafts are comparable to BMP-2 + ACS in spinal fusion efficacy. Histological differences were observed however, including robust endochondral ossification with NELL-1 treatment as compared to lipid-filled bone with BMP-2 treatment. These findings suggest NELL-1 based bone grafts show promise for future efforts in skeletal tissue engineering.


Assuntos
Proteína Morfogenética Óssea 2/uso terapêutico , Transplante Ósseo , Proteínas do Tecido Nervoso/uso terapêutico , Fusão Vertebral/métodos , Animais , Técnica de Desmineralização Óssea , Masculino , Ratos , Ratos Nus , Proteínas Recombinantes/uso terapêutico
7.
Tissue Eng Part A ; 19(11-12): 1386-97, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23406369

RESUMO

An ideal mesenchymal stem cell (MSC) source for bone tissue engineering has yet to be identified. Such an MSC population would be easily harvested in abundance, with minimal morbidity and with high purity. Our laboratories have identified perivascular stem cells (PSCs) as a candidate cell source. PSCs are readily isolatable through fluorescent-activated cell sorting from adipose tissue and have been previously shown to be indistinguishable from MSCs in the phenotype and differentiation potential. PSCs consist of two distinct cell populations: (1) pericytes (CD146+, CD34-, and CD45-), which surround capillaries and microvessels, and (2) adventitial cells (CD146-, CD34+, and CD45-), found within the tunica adventitia of large arteries and veins. We previously demonstrated the osteogenic potential of pericytes by examining pericytes derived from the human fetal pancreas, and illustrated their in vivo trophic and angiogenic effects. In the present study, we used an intramuscular ectopic bone model to develop the translational potential of our original findings using PSCs (as a combination of pericytes and adventitial cells) from human white adipose tissue. We evaluated human PSC (hPSC)-mediated bone formation and vascularization in vivo. We also examined the effects of hPSCs when combined with the novel craniosynostosis-associated protein, Nel-like molecule I (NELL-1). Implants consisting of the demineralized bone matrix putty combined with NELL-1 (3 µg/µL), hPSC (2.5×10(5) cells), or hPSC+NELL-1, were inserted in the bicep femoris of SCID mice. Bone growth was evaluated using microcomputed tomography, histology, and immunohistochemistry over 4 weeks. Results demonstrated the osteogenic potential of hPSCs and the additive effect of hPSC+NELL-1 on bone formation and vasculogenesis. Comparable osteogenesis was observed with NELL-1 as compared to the more commonly used bone morphogenetic protein-2. Next, hPSCs induced greater implant vascularization than the unsorted stromal vascular fraction from patient-matched samples. Finally, we observed an additive effect on implant vascularization with hPSC+NELL-1 by histomorphometry and immunohistochemistry, accompanied by in vitro elaboration of vasculogenic growth factors. These findings hold significant implications for the cell/protein combination therapy hPSC+NELL-1 in the development of strategies for vascularized bone regeneration.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/farmacologia , Osteogênese , Células-Tronco/citologia , Adulto , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Feminino , Humanos , Imuno-Histoquímica , Implantes Experimentais , Masculino , Camundongos , Camundongos SCID , Ovinos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos
8.
Tissue Eng Part A ; 19(3-4): 426-36, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23083222

RESUMO

Over 10 million Americans have osteoporosis, and is the predominant cause of fractures in the elderly. Treatment of fractures in the setting of osteoporosis is complicated by a suboptimal bone regenerative response due to a decline in the number of osteoblasts, their function, and survival. Consequently, an osteogenic therapeutic to prevent and treat fractures in patients with osteoporosis is needed. Nel-like molecule-1 (NELL-1), a novel osteoinductive growth factor, has been shown to promote bone regeneration. In this study, we aim to demonstrate the capacity of recombinant NELL-1 to prevent ovariectomy (OVX)-induced osteoporosis in a senile rat model. Ten-month-old female Sprague-Dawley rats underwent either sham surgery or OVX. Subsequently, 50 µL of 600 µg/mL NELL-1 lyophilized onto a 0-50-µm tricalcium phosphate (TCP) carrier was injected into the femoral bone marrow cavity while phosphate-buffered saline (PBS) control was injected into the contralateral femur. Our microcomputed tomography results showed that OVX+PBS/TCP control femurs showed a continuous decrease in the bone volume (BV) and bone mineral density (BMD) from 2 to 8 weeks post-OVX. In contrast, OVX+NELL-1/TCP femurs showed resistance to OVX-induced bone resorption showing BV and BMD levels similar to that of SHAM femurs at 8 weeks post-OVX. Histology showed increased endosteal-woven bone, as well as decreased adipocytes in the bone marrow of NELL-1-treated femurs compared to control. NELL-1-treated femurs also showed increased immunostaining for bone differentiation markers osteopontin and osteocalcin. These findings were validated in vitro, in which addition of NELL-1 in OVX bone marrow stem cells resulted in increased osteogenic differentiation. Thus, NELL-1 effectively enhances in situ osteogenesis in the bone marrow, making it potentially useful in the prevention and treatment of osteoporotic fractures.


Assuntos
Calcificação Fisiológica/efeitos dos fármacos , Fêmur/fisiopatologia , Proteínas do Tecido Nervoso/administração & dosagem , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Osteoporose/fisiopatologia , Animais , Feminino , Fêmur/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
9.
Stem Cells Transl Med ; 1(6): 510-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23197855

RESUMO

Adipose tissue is an ideal source of mesenchymal stem cells for bone tissue engineering: it is largely dispensable and readily accessible with minimal morbidity. However, the stromal vascular fraction (SVF) of adipose tissue is a heterogeneous cell population, which leads to unreliable bone formation. In the present study, we prospectively purified human perivascular stem cells (PSCs) from adipose tissue and compared their bone-forming capacity with that of traditionally derived SVF. PSCs are a population (sorted by fluorescence-activated cell sorting) of pericytes (CD146+CD34-CD45-) and adventitial cells (CD146-CD34+CD45-), each of which we have previously reported to have properties of mesenchymal stem cells. Here, we found that PSCs underwent osteogenic differentiation in vitro and formed bone after intramuscular implantation without the need for predifferentiation. We next sought to optimize PSCs for in vivo bone formation, adopting a demineralized bone matrix for osteoinduction and tricalcium phosphate particle formulation for protein release. Patient-matched, purified PSCs formed significantly more bone in comparison with traditionally derived SVF by all parameters. Recombinant bone morphogenetic protein 2 increased in vivo bone formation but with a massive adipogenic response. In contrast, recombinant Nel-like molecule 1 (NELL-1; a novel osteoinductive growth factor) selectively enhanced bone formation. These studies suggest that adipose-derived human PSCs are a new cell source for future efforts in skeletal regenerative medicine. Moreover, PSCs are a stem cell-based therapeutic that is readily approvable by the U.S. Food and Drug Administration, with potentially increased safety, purity, identity, potency, and efficacy. Finally, NELL-1 is a candidate growth factor able to induce human PSC osteogenesis.


Assuntos
Regeneração Óssea , Células-Tronco Mesenquimais/citologia , Osteogênese , Pericitos/citologia , Adipogenia , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Animais , Antígenos CD34/metabolismo , Matriz Óssea/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/farmacologia , Antígeno CD146/metabolismo , Fosfatos de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio , Técnicas de Cultura de Células , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Antígenos Comuns de Leucócito/metabolismo , Lipectomia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Proteínas do Tecido Nervoso/metabolismo , Pericitos/efeitos dos fármacos , Estudos Prospectivos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Medicina Regenerativa/métodos , Alicerces Teciduais , Microtomografia por Raio-X
10.
Stem Cells Transl Med ; 1(9): 673-84, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23197874

RESUMO

Adipose tissue is an ideal mesenchymal stem cell (MSC) source, as it is dispensable and accessible with minimal morbidity. However, the stromal vascular fraction (SVF) of adipose tissue is a heterogeneous cell population, which has disadvantages for tissue regeneration. In the present study, we prospectively purified human perivascular stem cells (PSCs) from n = 60 samples of human lipoaspirate and documented their frequency, viability, and variation with patient demographics. PSCs are a fluorescence-activated cell sorting-sorted population composed of pericytes (CD45-, CD146+, CD34-) and adventitial cells (CD45-, CD146-, CD34+), each of which we have previously reported to have properties of MSCs. Here, we found that PSCs make up, on average, 43.2% of SVF from human lipoaspirate (19.5% pericytes and 23.8% adventitial cells). These numbers were minimally changed by age, gender, or body mass index of the patient or by length of refrigerated storage time between liposuction and processing. In a previous publication, we observed that human PSCs (hPSCs) formed significantly more bone in vivo in comparison with unsorted human SVF (hSVF) in an intramuscular implantation model. We now extend this finding to a bone injury model, observing that purified hPSCs led to significantly greater healing of mouse critical-size calvarial defects than hSVF (60.9% healing as opposed to 15.4% healing at 2 weeks postoperative by microcomputed tomography analysis). These studies suggest that adipose-derived hPSCs are a new cell source for future efforts in skeletal regenerative medicine. Moreover, hPSCs are a stem cell-based therapeutic that is readily approvable by the U.S. Food and Drug Administration, with potentially increased safety, purity, identity, potency, and efficacy.


Assuntos
Regeneração Óssea , Osso e Ossos , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual , Tecido Adiposo/citologia , Túnica Adventícia , Animais , Antígenos CD34/análise , Antígeno CD146/análise , Separação Celular , Humanos , Antígenos Comuns de Leucócito/análise , Camundongos , Pericitos , Alicerces Teciduais , Cicatrização , Ferimentos e Lesões/terapia
11.
Biomaterials ; 33(34): 8745-56, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22959466

RESUMO

Implant-associated bacterial infections are one of the most serious complications in orthopedic surgery. Treatment of these infections often requires multiple operations, device removal, long-term systemic antibiotics, and extended rehabilitation, and is frequently ineffective, leading to worse clinical outcomes and increased financial costs. In this study, we evaluated silver nanoparticle/poly(DL-lactic-co-glycolic acid) (PLGA)-coated stainless steel alloy(SNPSA) as a potential antimicrobial implant material. We found that SNPSA exhibited strong antibacterial activity in vitro and ex vivo, and promoted MC3T3-E1 pre-osteoblasts proliferation and maturation in vitro. Furthermore, SNPSA implants induced osteogenesis while suppressing bacterial survival in contaminated rat femoral canals. Our results indicate that SNPSA has simultaneous antimicrobial and osteoinductive properties that make it a promising therapeutic material in orthopedic surgery.


Assuntos
Antibacterianos/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Osteogênese , Prata/farmacologia , Células 3T3 , Animais , Antibacterianos/química , Bactérias/efeitos dos fármacos , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/etiologia , Materiais Revestidos Biocompatíveis/química , Fêmur/diagnóstico por imagem , Fêmur/crescimento & desenvolvimento , Fêmur/microbiologia , Ácido Láctico/química , Masculino , Camundongos , Nanopartículas/química , Osteoblastos/citologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Próteses e Implantes/efeitos adversos , Radiografia , Ratos , Ratos Sprague-Dawley , Prata/química , Aço Inoxidável/química
12.
J Vis Exp ; (63): e2952, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22664543

RESUMO

Human perivascular stem cells (PSCs) can be isolated in sufficient numbers from multiple tissues for purposes of skeletal tissue engineering. PSCs are a FACS-sorted population of 'pericytes' (CD146+CD34-CD45-) and 'adventitial cells' (CD146-CD34+CD45-), each of which we have previously reported to have properties of mesenchymal stem cells. PSCs, like MSCs, are able to undergo osteogenic differentiation, as well as secrete pro-osteogenic cytokines. In the present protocol, we demonstrate the osteogenicity of PSCs in several animal models including a muscle pouch implantation in SCID (severe combined immunodeficient) mice, a SCID mouse calvarial defect and a femoral segmental defect (FSD) in athymic rats. The thigh muscle pouch model is used to assess ectopic bone formation. Calvarial defects are centered on the parietal bone and are standardly 4 mm in diameter (critically sized). FSDs are bicortical and are stabilized with a polyethylene bar and K-wires. The FSD described is also a critical size defect, which does not significantly heal on its own. In contrast, if stem cells or growth factors are added to the defect site, significant bone regeneration can be appreciated. The overall goal of PSC xenografting is to demonstrate the osteogenic capability of this cell type in both ectopic and orthotopic bone regeneration models.


Assuntos
Regeneração Óssea , Pericitos/citologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Fêmur/patologia , Humanos , Camundongos , Camundongos SCID , Modelos Animais , Ratos , Ratos Nus , Crânio/patologia , Alicerces Teciduais
13.
Biomaterials ; 33(24): 5821-31, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22622142

RESUMO

Pluripotent and/or multipotent stem cell-based therapeutics are a vital component of tissue engineering and regenerative medicine. The generation or isolation of safer and readily available stem cell sources will significantly aid clinical applications. We report here a technique using a single molecule, recombinant human fibromodulin protein (FMOD), to reprogram human fibroblasts into multipotent cells. Like virally-induced pluripotent stem (iPS) cells, FMOD reprogrammed (FReP) cells express pluripotency markers, form embryoid bodies (EBs), and differentiate into ectoderm, mesoderm, and endoderm derivatives in vitro. Notably, FReP cells regenerate muscle and bone tissues but do not generate teratomas in vivo. Unlike iPS cells, undifferentiated FReP cells proliferate slowly and express low proto-oncogene c-MYC and unexpectedly high levels of cyclin-dependent kinase inhibitors p15(Ink4B) and p21(WAF1/Cip1). Remarkably, in a fashion reminiscent of quiescent stem cells, the slow replicative phenotype of undifferentiated FReP cells reverses after differentiation induction, with differentiating FReP cells proliferating faster and expressing less p15(Ink4B) and p21(WAF1/Cip1) than differentiating iPS cells. Overall, single protein, FMOD-based, cell reprograming bypasses the risks of mutation, gene instability, and malignancy associated with genetically-modified iPS cells, and provides an alternative strategy for engineering patient-specific multipotent cells for basic research and therapeutic application.


Assuntos
Reprogramação Celular , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteoglicanas/metabolismo , Adulto , Osso e Ossos/citologia , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Fibromodulina , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , Músculo Esquelético/citologia , Proto-Oncogene Mas
14.
Stem Cells Dev ; 21(12): 2170-8, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22264144

RESUMO

A theoretical inverse relationship exists between osteogenic (bone forming) and adipogenic (fat forming) mesenchymal stem cell (MSC) differentiation. This inverse relationship in theory partially underlies the clinical entity of osteoporosis, in which marrow MSCs have a preference for adipose differentiation that increases with age. Two pro-osteogenic cytokines have been recently studied that each also possesses antiadipogenic properties: Sonic Hedgehog (SHH) and NELL-1 proteins. In the present study, we assayed the potential additive effects of the biologically active N-terminus of SHH (SHH-N) and NELL-1 protein on osteogenic and adipogenic differentiation of human primary adipose-derived stromal cell (hASCs). We observed that both recombinant SHH-N and NELL-1 protein significantly enhanced osteogenic differentiation and reduced adipose differentiation across all markers examined (alkaline phosphatase, Alizarin red and Oil red O staining, and osteogenic gene expression). Moreover, SHH-N and NELL-1 directed signaling produced additive effects on the pro-osteogenic and antiadipogenic differentiation of hASCs. NELL-1 treatment increased Hedgehog signaling pathway expression; coapplication of the Smoothened antagonist Cyclopamine reversed the pro-osteogenic effect of NELL-1. In summary, Hedgehog and Nell-1 signaling exert additive effects on the pro-osteogenic and antiadipogenic differentiation of ASCs. These studies suggest that the combination cytokines SHH-N+NELL-1 may represent a viable future technique for inducing the osteogenic differentiation of MSCs.


Assuntos
Adipogenia , Tecido Adiposo/citologia , Células-Tronco Adultas/fisiologia , Proteínas Hedgehog/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Osteogênese , Adulto , Células-Tronco Adultas/enzimologia , Células-Tronco Adultas/metabolismo , Fosfatase Alcalina/metabolismo , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Proteínas Hedgehog/farmacologia , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/farmacologia , Fenótipo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais , Receptor Smoothened , Alcaloides de Veratrum/farmacologia
15.
Tissue Eng Part A ; 18(3-4): 252-61, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21902605

RESUMO

Repair of cartilage due to joint trauma remains challenging due to the poor healing capacity of cartilage and adverse effects related to current growth factor-based strategies. NELL-1 (Nel-like molecule-1; Nel [a protein strongly expressed in neural tissue encoding epidermal growth factor like domain]), a protein first characterized in the context of premature cranial suture fusion, is believed to accelerate differentiation along the osteochondral lineage. We previously demonstrated the ability of NELL-1 protein to maintain the cartilaginous phenotype of explanted rabbit chondrocytes in vitro. Our objective in the current study is to determine whether NELL-1 can affect endogenous chondrocytes in an in vivo cartilage defect model. To generate the implant, NELL-1 was incorporated into chitosan nanoparticles and embedded into alginate hydrogels. These implants were press fit into 3-mm circular osteochondral defects created in the femoral condylar cartilage of 3-month-old New Zealand White rabbits (n=10). Controls included unfilled defects (n=8) and defects filled with phosphate-buffered saline-loaded chitosan nanoparticles embedded in alginate hydrogels (n=8). Rabbits were sacrificed 3 months postimplantation for histological analysis. Defects filled with alginate containing NELL-1 demonstrated significantly improved cartilage regeneration. Remarkably, histology of NELL-1-treated defects closely resembled that of native cartilage, including stronger Alcian blue and Safranin-O staining and increased deposition of type II collagen and absence of the bone markers type I collagen and Runt-related transcription factor 2 (Runx2) as demonstrated by immunohistochemistry. Our results suggest that NELL-1 may produce functional cartilage with properties similar to native cartilage, and is an exciting candidate for tissue engineering-based approaches for treating diverse pathologies of cartilage defects and degeneration.


Assuntos
Cartilagem/efeitos dos fármacos , Cartilagem/fisiologia , Proteínas do Tecido Nervoso/farmacologia , Alginatos/química , Animais , Células CHO , Proteínas de Ligação ao Cálcio , Cartilagem/patologia , Bovinos , Cricetinae , Cricetulus , Modelos Animais de Doenças , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Imuno-Histoquímica , Implantes Experimentais , Cinética , Coelhos , Regeneração , Coloração e Rotulagem
16.
PLoS One ; 6(9): e24638, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931789

RESUMO

NELL-1 is a novel secreted protein associated with premature fusion of cranial sutures in craniosynostosis that has been found to promote osteoblast cell differentiation and mineralization. Our previous study showed that Runx2, the key transcription factor in osteoblast differentiation, transactivates the NELL-1 promoter. In this study, we evaluated the regulatory involvement and mechanisms of Osterix, an essential transcription factor of osteoblasts, in NELL-1 gene expression and function. Promoter analysis showed a cluster of potential Sp1 sites (Sp1/Osterix binding sites) within approximately 70 bp (from -71 to -142) of the 5' flanking region of the human NELL-1 transcriptional start site. Luciferase activity in our NELL-1 promoter reporter systems was significantly decreased in Saos-2 cells when Osterix was overexpressed. Mutagenesis study demonstrated that this suppression is mediated by the Sp1 sites. The binding specificity of Osterix to these Sp1 sites was confirmed in Saos-2 cells and primary human osteoblasts by EMSA in vitro and ChIP assay in vivo. ChIP assay also showed that Osterix downregulated NELL-1 by affecting binding of RNA polymerase II to the NELL-1 promoter, but not by competing with Runx2 binding to the OSE2 sites. Moreover, NELL-1 mRNA levels were significantly decreased when Osterix was overexpressed in Saos-2, U2OS, Hela and Glioma cells. Correspondingly, knockdown of Osterix increased NELL-1 transcription and osteoblastic differentiation in both Saos-2 cells and primary human osteoblasts. These results suggest that Osterix is a direct transcriptional regulator with repressive effect on NELL-1 gene expression, contributing to a delicate balance of regulatory effects on NELL-1 transcription with Runx2, and may play a crucial role in osteoblast differentiation and mineralization. These findings also extend our understanding of the molecular mechanism of Runx2, Osterix, and NELL-1 and demonstrate their crosstalk during osteogenesis.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição/metabolismo , Sítios de Ligação/genética , Proteínas de Ligação ao Cálcio , Linhagem Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp7 , Fatores de Transcrição/genética
17.
Biochem Biophys Res Commun ; 411(1): 126-31, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21723263

RESUMO

A theoretical inverse relationship has long been postulated for osteogenic and adipogenic differentiation (bone versus adipose tissue differentiation). This inverse relationship in theory at least partially underlies the clinical entity of osteoporosis, in which marrow mesenchymal stem cells (MSCs) have a predilection for adipose differentiation that increases with age. In the present study, we assayed the potential anti-adipogenic effects of Nell-1 protein (an osteoinductive molecule). Using 3T3-L1 (a human preadipocyte cell line) cells and human adipose-derived stromal cells (ASCs), we observed that adenoviral delivered (Ad)-Nell-1 or recombinant NELL-1 protein significantly reduced adipose differentiation across all markers examined (Oil red O staining, adipogenic gene expression [Pparg, Lpl, Ap2]). In a prospective fashion, Hedgehog signaling was assayed as potentially downstream of Nell-1 signaling in regulating osteogenic over adipogenic differentiation. In comparison to Ad-LacZ control, Ad-Nell-1 increased expression of hedgehog signaling markers (Ihh, Gli1, Ptc1). These studies suggest that Nell-1 is a potent anti-adipogenic agent. Moreover, Nell-1 signaling may inhibit adipogenic differentiation via a Hedgehog dependent mechanism.


Assuntos
Adipogenia , Proteínas Hedgehog/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Proteínas de Ligação ao Cálcio , Regulação da Expressão Gênica , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/farmacologia
18.
FEBS Lett ; 585(15): 2410-8, 2011 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-21723284

RESUMO

Nel-like protein 1 (NELL-1) is an osteoinductive molecule associated with premature calvarial suture closure. Here we identified apoptosis related protein 3 (APR3), a membrane protein known as a proliferation suppressor, as a binding protein of NELL-1 by biopanning. NELL-1 and APR3 colocalized on the nuclear envelope of human osteoblasts. NELL-1 significantly inhibited proliferation of osteoblasts co-transfected with APR3 through further down-regulation of Cyclin D1. The co-expression of NELL-1 and APR3 enhanced Ocn and Bsp expression and mineralization. RNAi of APR3 significantly reduced the differentiation effect of NELL-1. These findings suggest that the effects of NELL-1 on osteoblastic differentiation and proliferation are partly through binding to APR3.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Diferenciação Celular , Proliferação de Células , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Ciclina D1/metabolismo , Humanos , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso/fisiologia , Biblioteca de Peptídeos , Ligação Proteica/fisiologia
19.
J Bone Miner Res ; 26(6): 1230-41, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21611965

RESUMO

Nell-1 is a growth factor required for normal skeletal development and expression of extracellular matrix proteins required for bone and cartilage cell differentiation. We identified the transcription factor nuclear factor of activated T cells (Nfatc2) as a primary response gene of Nell-1 through a microarray screen, with validation using real-time polymerase chain reaction (PCR). We investigated the effects of recombinant Nell-1 protein on the chondrogenic cell line ATDC5 and primary mouse chondrocytes. The osteochondral transcription factor Runx2 was investigated as a possible intermediary between Nell-1 and Nfatc2 using adenoviral overexpression of wild-type and dominant-negative Runx2. Nell-1 transiently induced both transcription and translation of Nfatc2, an effect inhibited by transduction of dominant-negative Runx2, suggesting that Runx2 was necessary for Nfatc2 induction. Differentiation assays revealed inhibitory effects of Nell-1 on ATDC5 cells. Although proliferation was unaffected, expression of chondrocyte-specific genes was decreased, and cartilage nodule formation and proteoglycan accumulation were suppressed. siRNA knockdown of Nfatc2 significantly reversed these inhibitory effects. To elucidate the relationship between Nell-1, Runx2, and Nfatc2 in vivo, their presence and distribution were visualized in femurs of wild-type and Nell1-deficient mice at both neonatal and various developmental stages using immunohistochemistry. All three proteins colocalized in the perichondrium of wild-type femurs but stained weakly or were completely absent in Nell1-deficient femurs at neonatal stages. Thus Nfatc2 likely plays an important role in Nell-1-mediated osteochondral differentiation in vitro and in vivo. To our knowledge, this is the first demonstration that Nfatc2 is a primary response gene of Nell-1.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Condrócitos/metabolismo , Condrogênese/genética , Glicoproteínas/metabolismo , Fatores de Transcrição NFATC/genética , Animais , Proteínas de Ligação ao Cálcio/deficiência , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fêmur/citologia , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/deficiência , Humanos , Imuno-Histoquímica , Camundongos , Proteínas do Tecido Nervoso/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
20.
Tissue Eng Part A ; 17(19-20): 2497-509, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21615216

RESUMO

The search for novel sources of stem cells other than bone marrow mesenchymal stem cells (MSCs) for bone regeneration and repair has been a critical endeavor. We previously established an effective protocol to homogeneously purify human pericytes from multiple fetal and adult tissues, including adipose, bone marrow, skeletal muscle, and pancreas, and identified pericytes as a primitive origin of human MSCs. In the present study, we further characterized the osteogenic potential of purified human pericytes combined with a novel osteoinductive growth factor, Nell-1. Purified pericytes grown on either standard culture ware or human cancellous bone chip (hCBC) scaffolds exhibited robust osteogenic differentiation in vitro. Using a nude mouse muscle pouch model, pericytes formed significant new bone in vivo as compared to scaffold alone (hCBC). Moreover, Nell-1 significantly increased pericyte osteogenic differentiation, both in vitro and in vivo. Interestingly, Nell-1 significantly induced pericyte proliferation and was observed to have pro-angiogenic effects, both in vitro and in vivo. These studies suggest that pericytes are a potential new cell source for future efforts in skeletal regenerative medicine, and that Nell-1 is a candidate growth factor able to induce pericyte osteogenic differentiation.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Osteogênese/efeitos dos fármacos , Pericitos/citologia , Pericitos/efeitos dos fármacos , Animais , Regeneração Óssea/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Proteínas de Ligação ao Cálcio , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Feto/citologia , Membro Posterior/diagnóstico por imagem , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Membro Posterior/cirurgia , Humanos , Imuno-Histoquímica , Implantes Experimentais , Camundongos , Camundongos SCID , Neovascularização Fisiológica/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Pâncreas/embriologia , Pericitos/metabolismo , Pericitos/transplante , Implantação de Prótese , Alicerces Teciduais , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...