Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 30(9): 1179-1198.e7, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37683603

RESUMO

Osteoarthritis is a degenerative joint disease that causes pain, degradation, and dysfunction. Excessive canonical Wnt signaling in osteoarthritis contributes to chondrocyte phenotypic instability and loss of cartilage homeostasis; however, the regulatory niche is unknown. Using the temporomandibular joint as a model in multiple species, we identify Lgr5-expressing secretory cells as forming a Wnt inhibitory niche that instruct Wnt-inactive chondroprogenitors to form the nascent synovial joint and regulate chondrocyte lineage and identity. Lgr5 ablation or suppression during joint development, aging, or osteoarthritis results in depletion of Wnt-inactive chondroprogenitors and a surge of Wnt-activated, phenotypically unstable chondrocytes with osteoblast-like properties. We recapitulate the cartilage niche and create StemJEL, an injectable hydrogel therapy combining hyaluronic acid and sclerostin. Local delivery of StemJEL to post-traumatic osteoarthritic jaw and knee joints in rabbit, rat, and mini-pig models restores cartilage homeostasis, chondrocyte identity, and joint function. We provide proof of principal that StemJEL preserves the chondrocyte niche and alleviates osteoarthritis.


Assuntos
Condrócitos , Osteoartrite , Suínos , Animais , Coelhos , Ratos , Porco Miniatura , Cartilagem , Envelhecimento , Receptores Acoplados a Proteínas G
2.
Orthod Craniofac Res ; 26 Suppl 1: 131-141, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36891610

RESUMO

OBJECTIVE: The temporomandibular joint (TMJ) is anatomically comprised of the mandibular condylar cartilage (CC) lined with fibrocartilaginous superficial zone and is crucial for eating and dental occlusion. TMJ osteoarthritis (OA) leads to pain, joint dysfunction and permanent loss of cartilage tissue. However, there are no drugs clinically available that ameliorate OA and little is known about global profiles of genes that contribute to TMJ OA. Furthermore, animal models that recapitulate the complexity of signalling pathways contributing to OA pathogenesis are crucial for designing novel biologics that thwart OA progression. We have previously developed a New Zealand white rabbit TMJ injury model that demonstrates CC degeneration. Here, we performed genome-wide profiling to identify new signalling pathways critical for cellular functions during OA pathology. MATERIALS AND METHODS: Temporomandibular joint OA was surgically induced in New Zealand white rabbits. Three months following injury, we performed global gene expression profiling of the TMJ condyle. RNA samples from TMJ condyles were subjected to sequencing. After raw RNA-seq data were mapped to relevant genomes, differential expression was analysed with DESeq2. Gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were conducted. RESULTS/CONCLUSIONS: Our study revealed multiple pathways altered during TMJ OA induction including the Wnt, Notch and PI3K-Akt signalling pathways. We demonstrate an animal model that recapitulates the complexity of the cues and signals underlying TMJ OA pathogenesis, which is essential for developing and testing novel pharmacologic agents to treat OA.


Assuntos
Cartilagem Articular , Osteoartrite , Coelhos , Animais , RNA-Seq , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Articulação Temporomandibular , Côndilo Mandibular/metabolismo , Cartilagem/metabolismo , Cartilagem/patologia , Osteoartrite/genética , Osteoartrite/metabolismo , Cartilagem Articular/metabolismo
3.
Acta Biomater ; 143: 26-38, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35292413

RESUMO

Over the past two decades in situ tissue engineering has emerged as a new approach where biomaterials are used to harness the body's own stem/progenitor cells to regenerate diseased or injured tissue. Immunomodulatory biomaterials are designed to promote a regenerative environment, recruit resident stem cells to diseased or injured tissue sites, and direct them towards tissue regeneration. This review explores advances gathered from in vitro and in vivo studies on in situ tissue regenerative therapies. Here we also examine the different ways this approach has been incorporated into biomaterial sciences in order to create customized biomaterial products for therapeutic applications in a broad spectrum of tissues and diseases. STATEMENT OF SIGNIFICANCE: Biomaterials can be designed to recruit stem cells and coordinate their behavior and function towards the restoration or replacement of damaged or diseased tissues in a process known as in situ tissue regeneration. Advanced biomaterial constructs with precise structure, composition, mechanical, and physical properties can be transplanted to tissue site and exploit local stem cells and their micro-environment to promote tissue regeneration. In the absence of cells, we explore the critical immunomodulatory, chemical and physical properties to consider in material design and choice. The application of biomaterials for in situ tissue regeneration has the potential to address a broad range of injuries and diseases.


Assuntos
Materiais Biocompatíveis , Engenharia Tecidual , Materiais Biocompatíveis/farmacologia , Células-Tronco , Cicatrização
4.
FASEB J ; 34(3): 4445-4461, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32030828

RESUMO

Temporomandibular joint osteoarthritis (TMJ OA) leads to permanent cartilage destruction, jaw dysfunction, and compromises the quality of life. However, the pathological mechanisms governing TMJ OA are poorly understood. Unlike appendicular articular cartilage, the TMJ has two distinct functions as the synovial joint of the craniofacial complex and also as the site for endochondral jaw bone growth. The established dogma of endochondral bone ossification is that hypertrophic chondrocytes undergo apoptosis, while invading vasculature with osteoprogenitors replace cartilage with bone. However, contemporary murine genetic studies support the direct differentiation of chondrocytes into osteoblasts and osteocytes in the TMJ. Here we sought to characterize putative vasculature and cartilage to bone transdifferentiation using healthy and diseased TMJ tissues from miniature pigs and humans. During endochondral ossification, the presence of fully formed vasculature expressing CD31+ endothelial cells and α-SMA+ vascular smooth muscle cells were detected within all cellular zones in growing miniature pigs. Arterial, endothelial, venous, angiogenic, and mural cell markers were significantly upregulated in miniature pig TMJ tissues relative to donor matched knee meniscus fibrocartilage tissue. Upon surgically creating TMJ OA in miniature pigs, we discovered increased vasculature and putative chondrocyte to osteoblast transformation dually marked by COL2 and BSP or RUNX2 within the vascular bundles. Pathological human TMJ tissues also exhibited increased vasculature, while isolated diseased human TMJ cells exhibited marked increased in vasculature markers relative to control 293T cells. Our study provides evidence to suggest that the TMJ in higher order species are in fact vascularized. There have been no reports of cartilage to bone transdifferentiation or vasculature in human-relevant TMJ OA large animal models or in human TMJ tissues and cells. Therefore, these findings may potentially alter the clinical management of TMJ OA by defining new drugs that target angiogenesis or block the cartilage to bone transformation.


Assuntos
Transdiferenciação Celular/fisiologia , Condrócitos/citologia , Osteoartrite/diagnóstico , Osteoartrite/terapia , Osteoblastos/citologia , Animais , Apoptose , Células Cultivadas , Condrócitos/metabolismo , Cães , Ensaio de Imunoadsorção Enzimática , Feminino , Células HEK293 , Humanos , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Masculino , Osteoartrite/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Software , Suínos , Transtornos da Articulação Temporomandibular/diagnóstico , Transtornos da Articulação Temporomandibular/metabolismo , Transtornos da Articulação Temporomandibular/terapia
5.
PLoS One ; 14(10): e0223244, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31603905

RESUMO

The temporomandibular joint (TMJ) is a fibrocartilaginous tissue critical for chewing and speaking. In patients with temporomandibular disorders (TMDs), permanent tissue loss can occur. Recapitulating the complexity of TMDs in animal models is difficult, yet critical for the advent of new therapies. Synovial fluid from diseased human samples revealed elevated levels of tumor necrosis factor alpha (TNF-alpha). Here, we propose to recapitulate these findings in mice by subjecting murine TMJs with TNF-alpha or CFA (Complete Freund's Adjuvant) in mandibular condyle explant cultures and by local delivery in vivo using TMJ intra-articular injections. Both TNF-alpha and CFA delivery to whole mandibular explants and in vivo increased extracellular matrix deposition and increased cartilage thickness, while TNF-alpha treated explants had increased expression of inflammatory cytokines and degradative enzymes. Moreover, the application of TNF-alpha or CFA in both models reduced cell number. CFA delivery in vivo caused soft tissue inflammation, including pannus formation. Our work provides two methods of chemically induced TMJ inflammatory arthritis through a condyle explant model and intra-articular injection model that replicate findings seen in synovial fluid of human patients, which can be used for further studies delineating the mechanisms underlying TMJ pathology.


Assuntos
Artrite Experimental/imunologia , Cartilagem Articular/imunologia , Matriz Extracelular/imunologia , Transtornos da Articulação Temporomandibular/imunologia , Articulação Temporomandibular/imunologia , Proteína ADAMTS5/genética , Proteína ADAMTS5/imunologia , Adolescente , Adulto , Idoso , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/genética , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Colágeno Tipo II/genética , Colágeno Tipo II/imunologia , Colágeno Tipo X/genética , Colágeno Tipo X/imunologia , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Feminino , Adjuvante de Freund/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Interleucinas/genética , Interleucinas/imunologia , Masculino , Côndilo Mandibular/efeitos dos fármacos , Côndilo Mandibular/imunologia , Côndilo Mandibular/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Líquido Sinovial/imunologia , Articulação Temporomandibular/efeitos dos fármacos , Articulação Temporomandibular/patologia , Transtornos da Articulação Temporomandibular/genética , Transtornos da Articulação Temporomandibular/patologia , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/administração & dosagem
6.
Nat Commun ; 7: 13073, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27721375

RESUMO

Tissue regeneration using stem cell-based transplantation faces many hurdles. Alternatively, therapeutically exploiting endogenous stem cells to regenerate injured or diseased tissue may circumvent these challenges. Here we show resident fibrocartilage stem cells (FCSCs) can be used to regenerate and repair cartilage. We identify FCSCs residing within the superficial zone niche in the temporomandibular joint (TMJ) condyle. A single FCSC spontaneously generates a cartilage anlage, remodels into bone and organizes a haematopoietic microenvironment. Wnt signals deplete the reservoir of FCSCs and cause cartilage degeneration. We also show that intra-articular treatment with the Wnt inhibitor sclerostin sustains the FCSC pool and regenerates cartilage in a TMJ injury model. We demonstrate the promise of exploiting resident FCSCs as a regenerative therapeutic strategy to substitute cell transplantation that could be beneficial for patients suffering from fibrocartilage injury and disease. These data prompt the examination of utilizing this strategy for other musculoskeletal tissues.


Assuntos
Fibrocartilagem/citologia , Regeneração , Transplante de Células-Tronco , Células-Tronco/citologia , Articulação Temporomandibular/patologia , Articulação Temporomandibular/fisiopatologia , Cicatrização , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso e Ossos/patologia , Diferenciação Celular , Condrócitos/patologia , Glicoproteínas/metabolismo , Homeostase , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos Nus , Modelos Biológicos , Coelhos , Ratos Sprague-Dawley , Nicho de Células-Tronco , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
7.
Front Oral Biol ; 18: 1-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26599112

RESUMO

The periodontal ligament (PDL) and alveolar bone are two critical tissues for understanding orthodontic tooth movement. The current literature is replete with descriptive studies of multiple cell types and their matrices in the PDL and alveolar bone, but is deficient with how stem/progenitor cells differentiate into PDL and alveolar bone cells. Can one type of orthodontic force with a specific magnitude and frequency activate osteoblasts, whereas another force type activates osteoclasts? This chapter will discuss the biology of not only mature cells and their matrices in the periodontal ligament and alveolar bone, but also stem/progenitor cells that differentiate into fibroblasts, osteoblasts and osteoclasts. Key advances in tooth movement rely on further understanding of osteoblast and fibroblast differentiation from mesenchymal stem/progenitor cells, and osteoclastogenesis from the hematopoietic/monocyte lineage.


Assuntos
Adaptação Fisiológica/fisiologia , Processo Alveolar/fisiologia , Ligamento Periodontal/fisiologia , Técnicas de Movimentação Dentária/métodos , Processo Alveolar/citologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fibroblastos/fisiologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Ligamento Periodontal/citologia
8.
Plast Reconstr Surg ; 133(6): 1344-1353, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24867716

RESUMO

BACKGROUND: Current augmentative and reconstructive rhinoplasties use auto logous tissue grafts or synthetic bioinert materials to repair nasal trauma or attain an aesthetic shape. Autologous grafts are associated with donor-site trauma and morbidity. Synthetic materials are widely used but often yield an unnatural appearance and are prone to infection or dislocation. There is an acute clinical need for the generation of native tissues to serve as rhinoplasty grafts without the undesirable features that are associated with autologous grafts or current synthetic materials. METHODS: Bioactive scaffolds were developed that not only recruited cells in the nasal dorsum in vivo, but also induced chondrogenesis of the recruited cells. Bilayered scaffolds were fabricated with alginate-containing gelatin microspheres encapsulating cytokines atop a porous poly(lactic-co-glycolic acid) base. Microspheres were fabricated to contain recombinant human transforming growth factor-ß3 at doses of 200, 500, or 1000 ng, with phosphate-buffered saline-loaded microspheres used as a control. A rat model of augmentation rhinoplasty was created by implanting scaffolds atop the native nasal cartilage surface that was scored to induce cell migration. Tissue formation and chondrogenesis in the scaffolds were evaluated by image analysis and histologic staining with hematoxylin and eosin, toluidine blue, Verhoeff elastic-van Geison, and aggrecan immunohistochemistry. RESULTS: Sustained release of increasing doses of transforming growth factor-ß3 for up to the tested 10 weeks promoted orthotopic cartilage-like tissue formation in a dose-dependent manner. CONCLUSIONS: These findings represent the first attempt to engineer cartilage tissue by cell homing for rhinoplasty, and could potentially serve as an alternative material for augmentative and reconstructive rhinoplasty.


Assuntos
Rinoplastia/métodos , Técnicas de Cultura de Tecidos/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Materiais Biocompatíveis/uso terapêutico , Movimento Celular , Condrogênese/fisiologia , Materiais Revestidos Biocompatíveis/química , Ácido Láctico/uso terapêutico , Células-Tronco Mesenquimais/citologia , Microesferas , Modelos Animais , Cartilagens Nasais/citologia , Ácido Poliglicólico/uso terapêutico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Procedimentos de Cirurgia Plástica , Fator de Crescimento Transformador beta3/administração & dosagem
9.
Biomaterials ; 35(7): 2172-80, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24345734

RESUMO

Rodent incisors provide a classic model for studying epithelial-mesenchymal interactions in development. However, postnatal stem/progenitor cells in rodent incisors have not been exploited for tooth regeneration. Here, we characterized postnatal rat incisor epithelium and mesenchyme stem/progenitor cells and found that they formed enamel- and dentin-like tissues in vivo. Epithelium and mesenchyme cells were harvested separately from the apical region of postnatal 4-5 day rat incisors. Epithelial and mesenchymal phenotypes were confirmed by immunocytochemistry, CFU assay and/or multi-lineage differentiation. CK14+, Sox2+ and Lgr5+ epithelium stem cells from the cervical loop enhanced amelogenin and ameloblastin expression upon BMP4 or FGF3 stimulation, signifying their differentiation towards ameloblast-like cells, whereas mesenchyme stem/progenitor cells upon BMP4, BMP7 and Wnt3a treatment robustly expressed Dspp, a hallmark of odontoblastic differentiation. We then control-released microencapsulated BMP4, BMP7 and Wnt3a in transplants of epithelium and mesenchyme stem/progenitor cells in the renal capsule of athymic mice in vivo. Enamel and dentin-like tissues were generated in two integrated layers with specific expression of amelogenin and ameloblastin in the newly formed, de novo enamel-like tissue, and DSP in dentin-like tissue. These findings suggest that postnatal epithelium and mesenchyme stem/progenitor cells can be primed towards bioengineered tooth regeneration.


Assuntos
Amelogênese , Dentinogênese , Células Epiteliais/citologia , Células-Tronco Mesenquimais/citologia , Animais , Camundongos , Camundongos Nus , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
10.
Endod Topics ; 28(1): 106-117, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24976816

RESUMO

The goal of regenerative endodontics is to restore the functions of the dental pulp-dentin complex. Two approaches are being applied toward dental pulp-dentin regeneration: cell transplantation and cell homing. The majority of previous approaches are based on cell transplantation by delivering ex vivo cultivated cells toward dental pulp or dentin regeneration. Many hurdles limit the clinical translation of cell transplantation such as the difficulty of acquiring and isolating viable cells, uncertainty of what cells or what fractions of cells to use, excessive cost of cell manipulation and transportation, and the risk of immune rejection, pathogen transmission, and tumorigenesis in associated with ex vivo cell manipulation. In contrast, cell homing relies on induced chemotaxis of endogenous cells and therefore circumvents many of the difficulties that are associated with cell transplantation. An array of proteins, peptides, and chemical compounds that are yet to be identified may orchestrate endogenous cells to regenerate dental pulp-dentin complex. Both cell transplantation and cell homing are scientifically valid approaches; however, cell homing offers a number of advantages that are compatible with the development of clinical therapies for dental pulp-dentin regeneration.

11.
Cell Tissue Res ; 347(3): 665-76, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22382390

RESUMO

From its inception, tissue engineering has had three tenets: cells, biomaterial scaffolds and signaling molecules. Among the triad, cells are the center piece, because cells are the building blocks of tissues. For decades, cell therapies have focused on the procurement, manipulation and delivery of healthy cells for the treatment of diseases or trauma. Given the complexity and potential high cost of cell delivery, there is recent and surging interest to orchestrate endogenous cells for tissue regeneration. Biomaterial scaffolds are vital for many but not all, tissue-engineering applications and serve to accommodate or promote multiple cellular functions. Signaling molecules can be produced by transplanted cells or endogenous cells, or delivered specifically to regulate cell functions. This review highlights recent work in tissue engineering and cell therapies, with a focus on harnessing the capacity of endogenous cells as an alternative or adjunctive approach for tissue regeneration.


Assuntos
Fenômenos Fisiológicos Musculoesqueléticos , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Humanos , Transplante de Células-Tronco
12.
Am J Dent ; 24(6): 331-5, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22263328

RESUMO

PURPOSE: To explore a relationship between the size of pulp chamber perforation and reparative dentin formation in a canine model. METHODS: Pulp defects were created in the pulp chambers of maxillary and mandibular premolars (N = 64) in 17 healthy mongrel dogs in three different sizes (diameter/depth: 1/1, 2/1, and 2/2 mm3) with sterile round burs under general anesthesia. The perforations were immediately capped with hard-setting calcium hydroxide (CH) in the control group or sealed with Teflon membrane (TM) in the experimental group, followed by restoration with reinforced zinc oxide eugenol cement in vivo. Seven and 30 days after pulp chamber perforation and restoration all treated and control premolars were extracted and prepared for histomorphometric and statistical analyses. RESULTS: Reparative dentin formation was more pronounced for defect sizes up to 2/1 mm3 when treated with CH, and completely bridged the surgically created dentin defects only after 30 days. However, reparative dentin upon CH treatment failed to completely bridge pulp chamber exposure for 2/2 defects. By contrast, TM treatment only yielded mild reparative dentin bridging for defects up to 1/1, but not for either 2/1 or 2/2 defects at 30 days. Inflammatory responses of the exposed dental pulp tissue were more robust with the TM group than with the CH group. Thus, dental pulp tissue possesses a capacity for spontaneous repair by the formation of reparative dentin in this preclinical model, but only up to a defect size of -2 mm in diameter and 1 mm in depth. All observations are based on 30 days post-treatment in the canine model. These findings may serve as baseline for regenerative endodontic studies.


Assuntos
Capeamento da Polpa Dentária/veterinária , Polpa Dentária/patologia , Modelos Animais de Doenças , Animais , Cães
13.
Am J Pathol ; 176(2): 812-26, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20035055

RESUMO

The temporomandibular joint is critical for jaw movements and allows for mastication, digestion of food, and speech. Temporomandibular joint osteoarthritis is a degenerative disease that is marked by permanent cartilage destruction and loss of extracellular matrix (ECM). To understand how the ECM regulates mandibular condylar chondrocyte (MCC) differentiation and function, we used a genetic mouse model of temporomandibular joint osteoarthritis that is deficient in two ECM proteins, biglycan and fibromodulin (Bgn(-/0)Fmod(-/-)). Given the unavailability of cell lines, we first isolated primary MCCs and found that they were phenotypically unique from hyaline articular chondrocytes isolated from the knee joint. Using Bgn(-/0) Fmod(-/-) MCCs, we discovered the early basis for temporomandibular joint osteoarthritis arises from abnormal and accelerated chondrogenesis. Transforming growth factor (TGF)-beta1 is a growth factor that is critical for chondrogenesis and binds to both biglycan and fibromodulin. Our studies revealed the sequestration of TGF-beta1 was decreased within the ECM of Bgn(-/0) Fmod(-/-) MCCs, leading to overactive TGF-beta1 signal transduction. Using an explant culture system, we found that overactive TGF-beta1 signals induced chondrogenesis and ECM turnover in this model. We demonstrated for the first time a comprehensive study revealing the importance of the ECM in maintaining the mandibular condylar cartilage integrity and identified biglycan and fibromodulin as novel key players in regulating chondrogenesis and ECM turnover during temoporomandibular joint osteoarthritis pathology.


Assuntos
Condrogênese/genética , Proteínas da Matriz Extracelular/fisiologia , Matriz Extracelular/metabolismo , Osteoartrite/genética , Proteoglicanas/fisiologia , Transtornos da Articulação Temporomandibular/genética , Articulação Temporomandibular/patologia , Animais , Biglicano , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/patologia , Matriz Extracelular/genética , Matriz Extracelular/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibromodulina , Masculino , Côndilo Mandibular/metabolismo , Côndilo Mandibular/patologia , Camundongos , Camundongos Knockout , Osteoartrite/metabolismo , Osteoartrite/patologia , Proteoglicanas/genética , Proteoglicanas/metabolismo , Articulação Temporomandibular/metabolismo , Transtornos da Articulação Temporomandibular/metabolismo , Transtornos da Articulação Temporomandibular/patologia
14.
Nat Med ; 13(10): 1219-27, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17828274

RESUMO

The repair of injured tendons remains a great challenge, largely owing to a lack of in-depth characterization of tendon cells and their precursors. We show that human and mouse tendons harbor a unique cell population, termed tendon stem/progenitor cells (TSPCs), that has universal stem cell characteristics such as clonogenicity, multipotency and self-renewal capacity. The isolated TSPCs could regenerate tendon-like tissues after extended expansion in vitro and transplantation in vivo. Moreover, we show that TSPCs reside within a unique niche predominantly comprised of an extracellular matrix, and we identify biglycan (Bgn) and fibromodulin (Fmod) as two critical components that organize this niche. Depletion of Bgn and Fmod affects the differentiation of TSPCs by modulating bone morphogenetic protein signaling and impairs tendon formation in vivo. Our results, while offering new insights into the biology of tendon cells, may assist in future strategies to treat tendon diseases.


Assuntos
Matriz Extracelular/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Tendões/citologia , Adipogenia , Animais , Biglicano , Diferenciação Celular , Separação Celular/métodos , Células Cultivadas , Criança , Condrogênese , Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibromodulina , Genes Reporter , Histocitoquímica , Humanos , Imuno-Histoquímica , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Osteogênese , Proteoglicanas/metabolismo , Transplante de Células-Tronco , Tendões/cirurgia , Transplante Homólogo
15.
Bone ; 40(4): 861-6, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17188951

RESUMO

Biglycan (Bgn) and decorin (Dcn) are highly expressed in numerous tissues in the craniofacial complex. However, their expression and function in the cranial sutures are unknown. In order to study this, we first examined the expression of biglycan and decorin in the posterior frontal suture (PFS), which predictably fuses between 21 and 45 days post-natal and in the non-fusing sagittal (S) suture from wild-type (Wt) mice. Our data showed that Bgn and Dcn were expressed in both cranial sutures. We then characterized the cranial suture phenotype in Bgn deficient, Dcn deficient, Bgn/Dcn double deficient, and Wt mice. At embryonic day 18.5, alizarin red/alcian blue staining showed that the Bgn/Dcn double deficient mice had hypomineralization of the frontal and parietal craniofacial bones. Histological analysis of adult mice (45-60 days post-natal) showed that the Bgn or Dcn deficient mice had no cranial suture abnormalities and immunohistochemistry staining showed increased production of Dcn in the PFS from Bgn deficient mice. To test possible compensation of Dcn in the Bgn deficient sutures, we examined the Bgn/Dcn double deficient mice and found that they had impaired fusion of the PFS. Semi-quantitative RT-PCR analysis of RNA from 35 day-old mice revealed increased expression of Bmp-4 and Dlx-5 in the PFS compared to their non-fusing S suture in Wt tissues and decreased expression of Dlx-5 in both PF and S sutures in the Bgn/Dcn double deficient mice compared to the Wt mice. Failure of PFS fusion and hypomineralization of the calvaria in the Bgn/Dcn double deficient mice demonstrates that these extracellular matrix proteoglycans could have a role in controlling the formation and growth of the cranial vault.


Assuntos
Suturas Cranianas/anormalidades , Proteínas da Matriz Extracelular/deficiência , Proteoglicanas/deficiência , Azul Alciano , Animais , Antraquinonas , Biglicano , Suturas Cranianas/embriologia , Suturas Cranianas/crescimento & desenvolvimento , Suturas Cranianas/metabolismo , Decorina , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Gravidez , Proteoglicanas/genética , Proteoglicanas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Coloração e Rotulagem
16.
Crit Rev Eukaryot Gene Expr ; 14(4): 301-15, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15663360

RESUMO

Biglycan is a member of the small leucine repeat proteoglycan family (SLRP). The biglycan gene is located on the X chromosome. Based on the amino acid sequence, the protein core of biglycan can be divided into six distinct domains: (1) a signal sequence, (2) a propeptide region, (3) a N-terminal glycosaminoglycan attachment region, (4) a cysteine loop, followed by (5) a leucine- rich repeat region domain (that makes up over 66% of the core protein), and (6) a final cysteine loop. Biglycan has been found in almost every organ within our body, but it is not uniformly distributed within an organ. Biglycan has been shown to be expressed on the cell surface, pericellularly, and sometimes within the extracellular matrices of a range of specialized cell types within the organ. Its expression pattern has been shown to be altered by growth factors and certain pathologic conditions. The regulation of biglycan expression occurs by both transcriptional and nontranscriptional mechanisms. The currently proposed biglycan functions appear to be dependent on the particular microenvironment and on the organ in question. In this review, we will focus on gene and protein structure, localization, expression, regulation, and function.


Assuntos
Proteoglicanas/fisiologia , Animais , Biglicano , Proteínas da Matriz Extracelular , Feminino , Expressão Gênica , Humanos , Masculino , Proteoglicanas/química , Proteoglicanas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA