Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Stem Cells ; 34(4): 1083-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26851078

RESUMO

Calcification of soft tissues, such as heart valves and tendons, is a common clinical problem with limited therapeutics. Tissue specific stem/progenitor cells proliferate to repopulate injured tissues. But some of them become divergent to the direction of ossification in the local pathological microenvironment, thereby representing a cellular target for pharmacological approach. We observed that HIF-2alpha (encoded by EPAS1 inclined form) signaling is markedly activated within stem/progenitor cells recruited at calcified sites of diseased human tendons and heart valves. Proinflammatory microenvironment, rather than hypoxia, is correlated with HIF-2alpha activation and promoted osteochondrogenic differentiation of tendon stem/progenitor cells (TSPCs). Abnormal upregulation of HIF-2alpha served as a key switch to direct TSPCs differentiation into osteochondral-lineage rather than teno-lineage. Notably, Scleraxis (Scx), an essential tendon specific transcription factor, was suppressed on constitutive activation of HIF-2alpha and mediated the effect of HIF-2alpha on TSPCs fate decision. Moreover, pharmacological inhibition of HIF-2alpha with digoxin, which is a widely utilized drug, can efficiently inhibit calcification and enhance tenogenesis in vitro and in the Achilles's tendinopathy model. Taken together, these findings reveal the significant role of the tissue stem/progenitor cells fate decision and suggest that pharmacological regulation of HIF-2alpha function is a promising approach for soft tissue calcification treatment.


Assuntos
Tendão do Calcâneo/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Calcinose/tratamento farmacológico , Terapia de Tecidos Moles , Tendão do Calcâneo/crescimento & desenvolvimento , Tendão do Calcâneo/patologia , Idoso , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Calcinose/genética , Calcinose/patologia , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Microambiente Celular/efeitos dos fármacos , Condrogênese/genética , Digoxina/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Cardiopatia Reumática/genética , Cardiopatia Reumática/patologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia
2.
Stem Cells ; 33(2): 443-55, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25332192

RESUMO

The transcription factor Mohawk (Mkx) is expressed in developing tendons and is an important regulator of tenogenic differentiation. However, the exact roles of Mkx in tendinopathy and tendon repair remain unclear. Using gene expression Omnibus datasets and immunofluorescence assays, we found that Mkx expression level was dramatically lower in human tendinopathy tissue and it is activated at specific stages of tendon development. In mesenchymal stem cells (MSCs), ectopic Mkx expression strikingly promoted tenogenesis more efficiently than Scleraxis (Scx), a well-known master transcription factor of tendon. Significantly higher levels of tenogenic gene expression and collagen fibril growth were observed with Mkx versus Scx. Interestingly, it was observed that Mkx dramatically upregulated Scx through binding to the Tgfb2 promoter. Additionally, the transplantation of Mkx-expressing-MSC sheets promoted tendon repair in a mouse model of Achilles-tendon defect. Taken together, these data shed light on previously unrecognized roles of Mkx in tendinopathy, tenogenesis, and tendon repair as well as in regulating the TGFß pathway.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Organogênese , Transdução de Sinais , Tendões/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Bases de Dados Genéticas , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Transgênicos , Tendinopatia/genética , Tendinopatia/metabolismo , Tendinopatia/patologia , Tendões/patologia , Fator de Crescimento Transformador beta2/genética
3.
Beijing Da Xue Xue Bao Yi Xue Ban ; 48(1): 1-4, 2016 Feb 18.
Artigo em Zh | MEDLINE | ID: mdl-26885900

RESUMO

The human embryonic stem cells (hESCs) serve as a self-renewable, genetically-healthy, pluripotent and single source of all body cells, tissues and organs. Therefore, it is considered as the good standard for all human stem cells by US, Europe and international authorities. In this study, the standard and healthy human mesenchymal progenitors, ligament tissues, cardiomyocytes, keratinocytes, primary neurons, fibroblasts, and salivary serous cells were differentiated from hESCs. The human cellular health-safety of NaF, retinoic acid, 5-fluorouracil, dexamethasone, penicillin G, adriamycin, lead acetate PbAc, bisphenol A-biglycidyl methacrylate (Bis-GMA) were evaluated selectively on the standardized platforms of hESCs, hESCs-derived cardiomyocytes, keratinocytes, primary neurons, and fibroblasts. The evaluations were compared with those on the currently most adopted cellular platforms. Particularly, the sensitivity difference of PM2.5 toxicity on standardized and healthy hESCs derived fibroblasts, currently adopted immortalized human bronchial epithelial cells Beas-2B and human umbilical vein endothelial cells (HUVECs) were evaluated. The RESULTS showed that the standardized hESCs cellular platforms provided more sensitivity and accuracy for human cellular health-safety evaluation.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Testes de Toxicidade , Diferenciação Celular , Fibroblastos/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Queratinócitos/citologia , Miócitos Cardíacos/citologia , Neurônios/citologia
4.
Ann Rheum Dis ; 74(1): 285-93, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24257023

RESUMO

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease characterised by cartilage degradation and chondrocyte hypertrophy. A recent study showed that Rac1 promoted expression of MMP13 and chondrocyte hypertrophy within the growth plate. These findings warrant further investigations on the roles of Rac1 in OA development and therapy in animal models. OBJECTIVE: To investigate the role and mechanistic pathway of Rac1 involvement in pathological changes of OA chondrocytes in vitro and OA development in vivo, as well as to develop a strategy of modulating Rac1 activity for OA treatment. MATERIAL AND METHODS: OA and normal cartilage from human or mice were used for immunohistochemical study and Rac1 activity assay. Chondrocytes treated with IL1ß and the untreated control were subjected to the Rac1 activity assay. Chondrocytes transfected with CA-Rac1, DN-Rac1 or GFP were cultured under conditions for inducing calcification. To evaluate the effect of Rac1 in OA development, an OA model was created by anterior cruciate ligament transection in mice. CA-Rac1, DN-Rac1 and GFP lentivirus, or NSC23766, were injected intra-articularly. Joints were subjected to histological analysis. RESULTS: It was found that there is aberrant Rac1 activation in human OA cartilage. Rac1 activity could also be elevated by IL1ß. Additionally, activated Rac1 promoted expression of MMP13, ADAMTS-5 and COLX by chondrocytes, partially through the ß-catenin pathway. Moreover, activation of Rac1 in knee joints by CA-Rac1 lentivirus accelerated OA progression, while inhibition of Rac1 activity by DN-Rac1 lentivirus or Rac1 inhibitor NSC23766 delayed OA development. Therefore, we developed a strategy of controlled release of NSC23766 from chitosan microspheres to OA joints, which effectively protected cartilage from destruction. CONCLUSIONS: These findings demonstrated that Rac1 activity is implicated in OA development. Also, controlled release of Rac1 inhibitor is a promising strategy for OA treatment.


Assuntos
Aminoquinolinas/farmacologia , Artrite Experimental/metabolismo , Calcinose/metabolismo , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Neuropeptídeos/metabolismo , Osteoartrite do Joelho/metabolismo , Pirimidinas/farmacologia , RNA Mensageiro/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS5 , Aminoquinolinas/administração & dosagem , Animais , Artrite Experimental/patologia , Artrite Experimental/terapia , Calcinose/patologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Quitosana , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Preparações de Ação Retardada , Perfilação da Expressão Gênica , Humanos , Hipertrofia , Metaloproteinase 13 da Matriz/genética , Camundongos , Microesferas , Neuropeptídeos/antagonistas & inibidores , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/terapia , Pirimidinas/administração & dosagem , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/genética
5.
Cell Tissue Res ; 360(2): 195-207, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25549759

RESUMO

Tendon injuries are commonly encountered in the clinic, disrupting the patient's normal work/life routine and damaging the career life of athletes. Currently, there is still no effective treatment for tendon injury. Tendon tissue engineering appears to be a promising route for tendon repair and regeneration. However, current strategies utilized in research are still far away from clinical applications due to unsuccessful cellular differentiation to tendon/tenocytes. In this review, we focus on the current physical strategies (mechanical stimulation and extracellular matrix topography) and evaluate their roles in precise and stepwise tendon differentiation. A systematic comprehension of normal tendon development process by structure, gene profile and physical microenvironment analysis is likely suggestive for stepwise tenocyte differentiation.


Assuntos
Diferenciação Celular , Células-Tronco/metabolismo , Traumatismos dos Tendões/terapia , Tendões/metabolismo , Engenharia Tecidual/métodos , Animais , Humanos , Células-Tronco/patologia , Traumatismos dos Tendões/metabolismo , Traumatismos dos Tendões/patologia , Tendões/patologia
6.
Beijing Da Xue Xue Bao Yi Xue Ban ; 45(1): 9-11, 2013 Feb 18.
Artigo em Zh | MEDLINE | ID: mdl-23411511

RESUMO

The current international standard for toxicity screening of biomedical devices and materials recommend the use of immortalized cell lines because of their homogeneous morphologies and infinite proliferation which provide good reproducibility for in vitro cytotoxicity screening. However, most of the widely used immortalized cell lines are derived from animals and may not be representative of normal human cell behavior in vivo, in particular in terms of the cytotoxic and genotoxic response. Therefore, It is vital to develop a model for toxicity evaluation. In our studies, two Chinese human embryonic stem cell (hESC) lines as toxicity model were established. hESC derived tissue/organ cell model for tissue/organ specific toxicity evaluation were developed. The efficiency and accuracy of using hESC model for cytoxicity, embryotoxicity and genotoxicity evaluation were confirmed. The results indicated that hESCs might be good tools for toxicity testing and biosafety evaluation in vitro.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Testes de Toxicidade/métodos , Povo Asiático , Técnicas de Cultura de Células , Avaliação Pré-Clínica de Medicamentos/métodos , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos
7.
Stem Cells ; 27(6): 1276-87, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489094

RESUMO

Human embryonic stem cells (hESCs) are ideal seed cells for tissue regeneration, but no research has yet been reported concerning their potential for tendon regeneration. This study investigated the strategy and efficacy of using hESCs for tendon regeneration as well as the mechanism involved. hESCs were first induced to differentiate into mesenchymal stem cells (MSCs), which had the potential to differentiate into the three mesenchymal lineages and were positive for MSC surface markers. hESC-derived MSCs (hESC-MSCs) regenerated tendon tissues in both an in vitro tissue engineering model and an in vivo ectopic tendon regeneration model, as confirmed by the expression of tendon-specific genes and structure. In in-situ rat patellar tendon repair, tendon treated with hESC-MSCs had much better structural and mechanical properties than did controls. Furthermore, hESC-MSCs remained viable at the tendon wound site for at least 4 weeks and secreted human fetal tendon-specific matrix components and differentiation factors, which then activated the endogenous regeneration process in tendon. Moreover, no teratoma was found in any samples. These findings demonstrate a safe and practical strategy of applying ESCs for tendon regeneration and may assist in future strategies to treat tendon diseases.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regeneração/fisiologia , Tendões/fisiologia , Engenharia Tecidual/métodos , Animais , Feminino , Citometria de Fluxo , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Traumatismos dos Tendões/cirurgia
8.
Cell Transplant ; 18(4): 433-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19622230

RESUMO

Mesenchymal stem cells (MSCs) hold great promise for bone regeneration. However, the power of mesenchymal stem cells has not been applied to structural bone allografts in clinical practice. This study designed a new strategy to enhance the efficiency of allografts for segmental bone regeneration. Isolated MSCs were cultured to form a cell sheet. The MSC sheet was then wrapped onto structural allografts. The assembled structures were cultured in vitro to evaluate the differentiation potential of MSC sheet. The assembled structures were implanted subcutaneously into nude mice as well as into the segmental radius defect of rabbits to investigate the efficiency of MSC sheets to repopulate allografts for bone repair. MSC sheets, upon assembling on bone grafts, showed similar differentiation properties to the in situ periosteum in vitro. After implantation the MSC sheets accelerated the repopulation of bone grafts in nude mice. Moreover, MSC sheets induced thicker cortical bone formation and more efficient graft-to-bone end fusion at the segmental bone defects in rabbits. This study thus presented a novel, more efficient, and practical strategy for large weight-bearing bone reconstruction by using MSC sheets to deliver large number of MSCs to repopulate the bone allografts.


Assuntos
Transplante Ósseo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Animais , Regeneração Óssea , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Nus , Osteogênese , Coelhos , Transplante Homólogo
9.
ACS Biomater Sci Eng ; 5(10): 5412-5421, 2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-33464061

RESUMO

Current surgical management of anterior cruciate ligament (ACL) rupture still remains an intractable challenge in ACL regeneration due to the weak self-healing capability of ACL. Inadequate cell numbers and vascularization within the articular cavity contribute mainly to the poor prognosis. This time, we fabricated a new tissue engineering scaffold by adding ligament stem/progenitor cell (LSPC) sheets to our previous knitted silk-collagen sponge scaffold, which overcame these limitations by providing sufficient numbers of seed cells and a natural extracellular matrix to facilitate regeneration. LSPCs display excellent proliferation and multilineage differentiation capacity. Upon ectopic implantation, the knitted silk-collagen sponge scaffold incorporated with an LSPC sheet exhibited less immune cells but more fibroblast-like cells, deposited ECM and neovascularization, and better tissue ingrowth. In a rabbit model, we excised the ACL and performed a reconstructive surgery with our scaffold. Increased expression of ligament-specific genes and better collagen fibril formation could be observed after orthotopic transplantation. After 6 months, the LSPC sheet group showed better results on ligament regeneration and ligament-bone healing. Furthermore, no obvious cartilage and meniscus degeneration were observed at 6 months postoperation. In conclusion, these results indicated that the new tissue engineering scaffold can promote ACL regeneration and slow down the progression of osteoarthritis, thus suggesting its high clinical potential as an ideal graft in ACL reconstruction.

10.
Cell Death Dis ; 10(6): 427, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31160550

RESUMO

Characterized by their slow adhering property, skeletal muscle myogenic progenitor cells (MPCs) have been widely utilized in skeletal muscle tissue engineering for muscle regeneration, but with limited efficacy. Skeletal muscle regeneration is regulated by various cell types, including a large number of rapidly adhering cells (RACs) where their functions and mechanisms are still unclear. In this study, we explored the function of RACs by co-culturing them with MPCs in a biomimetic skeletal muscle organoid system. Results showed that RACs promoted the myogenic potential of MPCs in the organoid. Single-cell RNA-Seq was also performed, classifying RACs into 7 cell subtypes, including one newly described cell subtype: teno-muscular cells (TMCs). Connectivity map of RACs and MPCs subpopulations revealed potential growth factors (VEGFA and HBEGF) and extracellular matrix (ECM) proteins involvement in the promotion of myogenesis of MPCs during muscle organoid formation. Finally, trans-well experiments and small molecular inhibitors blocking experiments confirmed the role of RACs in the promotion of myogenic differentiation of MPCs. The RACs reported here revealed complex cell diversity and connectivity with MPCs in the biomimetic skeletal muscle organoid system, which not only offers an attractive alternative for disease modeling and in vitro drug screening but also provides clues for in vivo muscle regeneration.


Assuntos
Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Organoides/citologia , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Análise por Conglomerados , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Mioblastos/citologia , Organoides/ultraestrutura , RNA-Seq , Análise de Célula Única , Transcriptoma/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Clin Invest ; 129(3): 1076-1093, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530994

RESUMO

Joint pain is the defining symptom of osteoarthritis (OA) but its origin and mechanisms remain unclear. Here, we investigated an unprecedented role of osteoclast-initiated subchondral bone remodeling in sensory innervation for OA pain. We show that osteoclasts secrete netrin-1 to induce sensory nerve axonal growth in subchondral bone. Reduction of osteoclast formation by knockout of receptor activator of nuclear factor kappa-B ligand (Rankl) in osteocytes inhibited the growth of sensory nerves into subchondral bone, dorsal root ganglion neuron hyperexcitability, and behavioral measures of pain hypersensitivity in OA mice. Moreover, we demonstrated a possible role for netrin-1 secreted by osteoclasts during aberrant subchondral bone remodeling in inducing sensory innervation and OA pain through its receptor DCC (deleted in colorectal cancer). Importantly, knockout of Netrin1 in tartrate-resistant acid phosphatase-positive (TRAP-positive) osteoclasts or knockdown of Dcc reduces OA pain behavior. In particular, inhibition of osteoclast activity by alendronate modifies aberrant subchondral bone remodeling and reduces innervation and pain behavior at the early stage of OA. These results suggest that intervention of the axonal guidance molecules (e.g., netrin-1) derived from aberrant subchondral bone remodeling may have therapeutic potential for OA pain.


Assuntos
Gânglios Espinais/metabolismo , Netrina-1/metabolismo , Osteoartrite/metabolismo , Osteoclastos/metabolismo , Dor/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Remodelação Óssea/genética , Receptor DCC/genética , Receptor DCC/metabolismo , Gânglios Espinais/patologia , Masculino , Camundongos , Netrina-1/genética , Osteoartrite/genética , Osteoartrite/patologia , Osteoclastos/patologia , Dor/genética , Dor/patologia , Células Receptoras Sensoriais/patologia
12.
Biomaterials ; 29(27): 3683-3692, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18541295

RESUMO

This study was aimed to develop a new practical ligament scaffold by synergistic incorporation of silk fibers, a knitted structure, and a collagen matrix. The efficacy for ligament tissue engineering was investigated in vitro and in animal models. Cells cultured on a collagen substrate expressed ligament matrix genes at higher levels than those on a silk substrate. The silk scaffold elicited little inflammatory reaction and degraded slowly after subcutaneous implantation in a mouse model. In the rabbit MCL defect model, MCLs treated with a silk+collagen scaffold deposited more collagen, had better mechanical properties, and showed more native microstructure with larger diameter collagen fibrils and stronger scaffold-ligament interface healing than untreated MCLs and those treated with silk scaffolds. These results demonstrated that the knitted silk+collagen sponge scaffold improves structural and functional ligament repair by regulating ligament matrix gene expression and collagen fibril assembly. The findings are the first to highlight the important roles of biomaterials in ligament regeneration biology. Also, the concept of an "internal-space-preservation" scaffold is proposed for the tissue repair under physical loading.


Assuntos
Colágeno , Ligamentos/fisiologia , Seda , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Feminino , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Coelhos , Regeneração , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Stem Cells Transl Med ; 7(5): 404-414, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29573225

RESUMO

Tendon disorders, which are commonly presented in the clinical setting, disrupt the patients' normal work and life routines, and they damage the careers of athletes. However, there is still no effective treatment for tendon disorders. In the field of tissue engineering, the potential of the therapeutic application of exogenous stem cells to treat tendon pathology has been demonstrated to be promising. With the development of stem cell biology and chemical biology, strategies that use inductive tenogenic factors to program stem cell fate in situ are the most easily and readily translatable to clinical applications. In this review, we focus on bioactive molecules that can potentially induce tenogenesis in adult stem cells, and we summarize the various differentiation factors found in comparative studies. Moreover, we discuss the molecular regulatory mechanisms of tenogenesis, and we examine the various challenges in developing standardized protocols for achieving efficient and reproducible tenogenesis. Finally, we discuss and predict future directions for tendon regeneration. Stem Cells Translational Medicine 2018;7:404-414.


Assuntos
Regeneração/fisiologia , Células-Tronco/citologia , Tendões/fisiologia , Animais , Diferenciação Celular/fisiologia , Humanos , Engenharia Tecidual/métodos , Alicerces Teciduais
14.
Curr Gene Ther ; 18(1): 29-39, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29651947

RESUMO

INTRODUCTION: Mesenchymal Stem Cells (MSCs) are promising candidates for nerve tissue engineering. Brain Derived Neurotrophic Factor (BDNF) secreted by MSCs can function to increase neural differentiation and relieve inflammation response. Gene transfection technology is an efficient strategy to increase the secretion levels of cytokines and enhance cellular functions. However, transfection and in vivo gene expression of environmentally sensitive stem cells have been one of the most challenging subjects due to the requirement in both safety and transfection efficiency. In this study, gene transfection technology was applied to prepare BDNF gene recombinant MSCs based on our previously reported liposomal vector ScreenFect® A. To improve cellular survival and gene expression after in situ implantation of MSCs, an adhesive peptide modified hydrogel scaffold was constructed using hyaluronic acid. The scaffold was optimized and modified with an adhesive peptide PPFLMLLKGSTR. The transfected MSCs exhibited improved cellular survival and sustained gene expression in the three-Dimentional (3D) scaffold in vitro. Compared to untransfected MSCs, gene recombinant MSCs effectively improved spinal tissue integrity, inhibited glial scar formation and alleviated inflammatory response. These effects were found discounted when cells were implanted without the scaffold. CONCLUSION: The study developed a promising implantation system for therapy of severe spinal cord injury and provided the first understanding of Screenfect® A about its functions on stem cell therapy for nerve tissue repair as well as three-dimentional gene expression.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Terapia Genética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Traumatismos da Medula Espinal/terapia , Adesivos/química , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Humanos , Hidrogéis/química , Masculino , Peptídeos/química , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/genética , Alicerces Teciduais/química , Transfecção
15.
Biomaterials ; 175: 44-60, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29800757

RESUMO

Great effort has been spent to promote the vascularization of tissue engineering bone grafts (TEBG) for improved therapeutic outcome. However, the thorough vascularization especially in the central region still remained as a major challenge for the clinical translation of TEBG. Here, we developed a new strategy to construct a centrally vascularized TEBG (CV-TEBG) with unique core-shell composite structure, which is consisted of an angiogenic core and an osteogenic shell. The in vivo evaluation in rabbit critical sized femoral defect was conducted to meticulously compare CV-TEBG to other TEBG designs (TEBG with osteogenic shell alone, or angiogenic core alone or angiogenic core+shell). Microfil-enhanced micro-CT analysis has been shown that CV-TEBG could outperform TEBG with pure osteogenic or angiogenic component for neo-vascularization. CV-TEBG achieved a much higher and more homogenous vascularization throughout the whole scaffold (1.52-38.91 folds, p < 0.01), and generated a unique burrito-like vascular network structure to perfuse both the central and peripheral regions of TEBG, indicating a potential synergistic effect between the osteogenic shell and angiogenic core in CV-TEBG to enhance neo-vascularization. Moreover, CV-TEBG has generated more new bone tissue than other groups (1.99-83.50 folds, p < 0.01), achieved successful bridging defect with the formation of both cortical bone like tissue externally and cancellous bone like tissue internally, and restored approximately 80% of the stiffness of the defected femur (benchmarked to the intact femur). It has been further observed that different bone regeneration patterns occurred in different TEBG implants and closely related to their vascularization patterns, revealing the potential profound influence of vascularization patterns on the osteogenesis pattern during defect healing.


Assuntos
Regeneração Óssea , Fêmur/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Alicerces Teciduais/química , Animais , Adesão Celular , Linhagem Celular , Proliferação de Células , Células Endoteliais/citologia , Fêmur/patologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Nus , Osteogênese , Polimetil Metacrilato/química , Coelhos , Engenharia Tecidual/métodos
16.
Acta Biomater ; 71: 168-183, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29524675

RESUMO

Anterior cruciate ligament (ACL) is one of the most difficult tissues to heal once injured. Ligament regeneration and tendon-bone junction healing are two major goals of ACL reconstruction. This study aimed to investigate the synergistic therapeutic effects of Stromal cell-derived factor 1 (SDF-1)-releasing collagen-silk (CSF) scaffold combined with intra-articular injection of ligament-derived stem/progenitor cells (LSPCs) for ACL regeneration and the amelioration in the long-term complication of osteoarthritis (OA). The stem cell recruitment ability of CSF scaffold and the multipotency, particularly the tendon forming ability of LSPCs from rabbits were characterized in vitro, while the synergistic effect of the CSF scaffold and LSPCs for ACL regeneration and OA amelioration were investigated in vivo at 1, 3, and 6 months with a rabbit ACL reconstruction model. The CSF scaffold was used as a substitute for the ACL, and LSPCs were injected into the joint cavity after 7 days of the ACL reconstruction. CSF scaffold displayed a controlled release pattern for the encapsulated protein for up to 7 days with an increased stiffness in the mechanical property. LSPCs, which exhibited highly I Collagen and CXCR4 expression, were attracted by SDF-1 and successfully relocated into the CSF scaffold at 1 month in vivo. At 3 and 6 months post-treatment, the CSF scaffold combined with LSPCs (CSFL group) enhanced the regeneration of ACL tissue, and promoted bone tunnel healing. Furthermore, the OA progression was impeded efficiently. Our findings here provided a new strategy that using stem cell recruiting CSF scaffold with tissue-specific stem cells, could be a promising solution for ACL regeneration. STATEMENT OF SIGNIFICANCE: In this study, we developed a silk scaffold with increased stiffness and SDF-1 controlled release capacity for ligament repair. This advanced scaffold transplantation combined with intra-articular injection of LSPCs (which was isolated from rabbit ligament for the first time in this study) promoted the regeneration of both the tendinous and bone tunnel portion of ACL. This therapeutic strategy also ameliorated cartilage degeneration and reduced the severity of arthrofibrosis. Hence, combining LSPCs injection with SDF-1-releasing silk scaffold is demonstrated as a therapeutic strategy for ACL regeneration and OA treatment in the clinic.


Assuntos
Ligamento Cruzado Anterior/metabolismo , Regeneração Óssea/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Fibroínas , Osteoartrite do Joelho/terapia , Transplante de Células-Tronco , Alicerces Teciduais/química , Animais , Ligamento Cruzado Anterior/patologia , Modelos Animais de Doenças , Fibroínas/química , Fibroínas/farmacologia , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Coelhos
17.
Biomaterials ; 28(28): 4056-67, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17590431

RESUMO

In this study a 3-D alginate microbead platform was coated with cartilaginous extracellular matrix (ECM) components to emulate chondrogenic microenvironment in vivo for the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). BMSCs were seeded onto the microbead surface and the effect of the modified microbead on BMSC adhesion, proliferation and chondrogenic differentiation was studied, and compared to chondrogenesis in conventional pellet culture. Our results indicated that microbead system promoted BMSC proliferation and protein deposition resulting in the formation of bigger aggregates compared to conventional pellet culture. Analysis of the aggregates indicated that chondroitin sulfate (CS)- and Col2-coated microbeads enhanced the chondrogenic differentiation of hBMSCs, with increasing formation of glycosaminoglycan (GAG) and collagen II deposition in histology, immunohistochemistry and real time PCR analysis. In addition, Col2-coated microbeads resulted in hypertrophic maturation of the differentiated chondrocytes, similar to conventional pellet culture, while CS-coated microbeads were able to retain the pre-hypertrophy state of the differentiated cells. Our result suggested that provision of suitable cartilaginous microenvironment in a 3-D system can promote the chondrogenic differentiation of BMSC and influence the phenotype of resulting chondrocytes. Our microbead system provides an easy method of processing a 3-D alginate system that allows the possibility of scaling up chondrogenic pellet production for clinical application, while the modifiable microbeads also provide an adjustable 3-D platform for the study of co-interaction of ECM and differentiation factors during the stem cell differentiation.


Assuntos
Cartilagem/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Matriz Extracelular/química , Células-Tronco Mesenquimais/fisiologia , Microesferas , Alginatos/química , Animais , Proliferação de Células , Células Cultivadas , Quitosana/química , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Propriedades de Superfície
18.
Tissue Eng ; 13(4): 659-66, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17371203

RESUMO

Cartilage damage has been documented as one of the major problems leading to knee repair procedures worldwide. The low availability of cartilage that can be harvested without causing a negative health impact has led to the focus on the potential of stem cells, which have been transplanted into damaged areas and successfully grown into new healthy tissue. This study aims to compare the chondrogenic potential of two stem cell sources--adipose tissue and bone marrow. Stem cells were isolated from donor-matched adipose tissue and bone marrow, following established protocols. The cells were grown in a chondrogenic cocktail containing transforming growth factor-beta3 (TGF-beta3) up till 28 days, and assessed for expression changes of cartilage markers at the gene and protein level, using qualitative and quantitative methods. Controls were included for every time point. Real-time polymerase chain reaction (PCR) results showed increases in the gene expression of collagen II in both the cell types that received TGF-beta3 treatment. However, histological, immunohistochemical, and glycosaminoglycan (GAG) assay clearly showed that collagen II and proteoglycans (PG) were synthesized only in the growth factor-treated bone marrow stem cells (BMSCs). These findings support the results obtained in our in vivo comparative study done on an animal model, suggesting that BMSCs are more suitable than adipose-derived stem cells (ADSCs) for chondrogenesis.


Assuntos
Adipócitos/citologia , Células da Medula Óssea/citologia , Condrócitos/citologia , Condrogênese/fisiologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Adolescente , Células da Medula Óssea/fisiologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Criança , Condrócitos/fisiologia , Feminino , Humanos , Masculino , Células-Tronco/fisiologia , Doadores de Tecidos
19.
Br J Oral Maxillofac Surg ; 45(4): 272-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17097777

RESUMO

The purpose of the current study is to fabricate tissue engineered trachea with poly-lactic-glycolic acid (PLGA) non-woven mesh enforced by collagen type I. PLGA fibres coated with collagen solution were put together and fabricated into the shape of a human trachea, after drying and cross-linking treatment, a non-woven mesh with "C" shape formed. Chondrocytes from sheep nasal septum cartilage were expanded in vitro and seeded into PLGA/collagen non-woven mesh in the density of 5.0 x 10(7)mL(-1). After 5 days of in vitro incubation, six Cell-PLGA/collagen composites were implanted subcutaneously into the back of 6 nude mice to prefabricate a tissue engineering trachea. Eight weeks later, the cartilage formation was observed by gross inspection and histological examination. Cartilage-like tissue in the shape of the initial PLGA/collagen scaffold had been regenerated successfully without obvious inflammatory response. The tissue engineered trachea cartilage consisted of evenly spaced lacunae embedded in matrix stained red with safranin-O staining. The amount of GAGs in tissue engineered trachea cartilage reached 71.42% of normal value in native cartilage. This study demonstrated that collagen-enforced PLGA non-woven mesh facilitated the adhesion and proliferation of chondrocytes, it also owned adequate mechanical strength to serve as an ideal scaffold for trachea tissue engineering without internal support.


Assuntos
Materiais Biocompatíveis , Colágeno Tipo I , Ácido Láctico , Ácido Poliglicólico , Polímeros , Telas Cirúrgicas , Engenharia Tecidual , Traqueia/anatomia & histologia , Animais , Materiais Biocompatíveis/química , Cartilagem/anatomia & histologia , Adesão Celular , Contagem de Células , Proliferação de Células , Células Cultivadas , Condrócitos/citologia , Condrogênese/fisiologia , Colágeno Tipo I/química , Corantes , Reagentes de Ligações Cruzadas/química , Dessecação , Glicosaminoglicanos/análise , Humanos , Ácido Láctico/química , Camundongos , Camundongos Nus , Septo Nasal/citologia , Fenazinas , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química , Ovinos , Tela Subcutânea/cirurgia , Engenharia Tecidual/métodos
20.
Acta Biomater ; 63: 64-75, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28890259

RESUMO

The demand of favorable scaffolds has increased for the emerging cartilage tissue engineering. Chondroitin sulfate (CS) and silk fibroin have been investigated and reported with safety and excellent biocompatibility as tissue engineering scaffolds. However, the rapid degradation rate of pure CS scaffolds presents a challenge to effectively recreate neo-tissue similar to natural articular cartilage. Meanwhile the silk fibroin is well used as a structural constituent material because its remarkable mechanical properties, long-lasting in vivo stability and hypoimmunity. The application of composite silk fibroin and CS scaffolds for joint cartilage repair has not been well studied. Here we report that the combination of silk fibroin and CS could synergistically promote articular cartilage defect repair. The silk fibroin (silk) and silk fibroin/CS (silk-CS) scaffolds were fabricated with salt-leaching, freeze-drying and crosslinking methodologies. The biocompatibility of the scaffolds was investigated in vitro by cell adhesion, proliferation and migration with human articular chondrocytes. We found that silk-CS scaffold maintained better chondrocyte phenotype than silk scaffold; moreover, the silk-CS scaffolds reduced chondrocyte inflammatory response that was induced by interleukin (IL)-1ß, which is in consistent with the well-documented anti-inflammatory activities of CS. The in vivo cartilage repair was evaluated with a rabbit osteochondral defect model. Silk-CS scaffold induced more neo-tissue formation and better structural restoration than silk scaffold after 6 and 12weeks of implantation in ICRS histological evaluations. In conclusion, we have developed a silk fibroin/ chondroitin sulfate scaffold for cartilage tissue engineering that exhibits immuno-inhibition property and can improve the self-repair capacity of cartilage. STATEMENT OF SIGNIFICANCE: Severe cartilage defect such as osteoarthritis (OA) is difficult to self-repair because of its avascular, aneural and alymphatic nature. Current scaffolds often focus on providing sufficient mechanical support or bio-mimetic structure to promote cartilage repair. Thus, silk has been adopted and investigated broadly. However, inflammation is one of the most important factors in OA. But few scaffolds for cartilage repair reported anti-inflammation property. Meanwhile, chondroitin sulfate (CS) is a glycosaminoglycan present in the natural cartilage ECM, and has exhibited a number of useful biological properties including anti-inflammatory activity. Thus, we designed this silk-CS scaffold and proved that this scaffold exhibited good anti-inflammatory effects both in vitro and in vivo, promoted the repair of articular cartilage defect in animal model.


Assuntos
Cartilagem Articular/fisiologia , Sulfatos de Condroitina/farmacologia , Fibroínas/farmacologia , Regeneração/efeitos dos fármacos , Alicerces Teciduais/química , Animais , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Cartilagem Articular/cirurgia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibroínas/ultraestrutura , Humanos , Interleucina-1beta/farmacologia , Masculino , Fenótipo , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA