Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
FASEB J ; 29(10): 4107-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26092928

RESUMO

We investigated the role of periostin, an extracellular matrix protein, in the pathophysiology of osteoarthritis (OA). In OA, dysregulated gene expression and phenotypic changes in articular chondrocytes culminate in progressive loss of cartilage from the joint surface. The molecular mechanisms underlying this process are poorly understood. We examined periostin expression by immunohistochemical analysis of lesional and nonlesional cartilage from human and rodent OA knee cartilage. In addition, we used small interfering (si)RNA and adenovirus transduction of chondrocytes to knock down and up-regulate periostin levels, respectively, and analyzed its effect on matrix metalloproteinase (MMP)-13, a disintegrin and MMP with thrombospondin motifs (ADAMTS)-4, and type II collagen expression. We found high periostin levels in human and rodent OA cartilage. Periostin increased MMP-13 expression dose [1-10 µg/ml (EC50 0.5-1 µg/ml)] and time (24-72 h) dependently, significantly enhanced expression of ADAMTS4 mRNA, and promoted cartilage degeneration through collagen and proteoglycan degradation. Periostin induction of MMP-13 expression was inhibited by CCT031374 hydrobromide, an inhibitor of the canonical Wnt/ß-catenin signaling pathway. In addition, siRNA-mediated knockdown of endogenous periostin blocked constitutive MMP-13 expression. These findings implicate periostin as a catabolic protein that promotes cartilage degeneration in OA by up-regulating MMP-13 through canonical Wnt signaling.


Assuntos
Cartilagem Articular/metabolismo , Moléculas de Adesão Celular/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Osteoartrite/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Bovinos , Moléculas de Adesão Celular/genética , Células Cultivadas , Condrócitos/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 13 da Matriz/genética , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Osteoartrite/genética , Pró-Colágeno N-Endopeptidase/genética , Pró-Colágeno N-Endopeptidase/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
ACS Biomater Sci Eng ; 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39259932

RESUMO

The secretion rate of albumin is a key indicator of function in liver tissue models used for hepatotoxicity and pharmacokinetic testing. However, it is not generally clear how to determine molecular secretion rates from measurements of the molecular concentration in supernatant media. Here, we develop computational and analytical models of molecular transport in an experimental system that enable determination of albumin secretion rates based on measurements of albumin concentration in supernatant media. The experimental system is a 3D-bioprinted human liver tissue construct embedded in a 3D culture environment made from packed microgel particles swollen in liquid growth media. The mathematical models reveal that the range of albumin synthesis rates necessary to match experimentally measured albumin concentrations corresponds to reaction-limited conditions, where a steady state of albumin spatial distribution is rapidly reached between media exchanges. Our results show that temporally resolved synthesis rates can be inferred from serial concentration measurements of collected supernatant media. This link is critical to confidently assessing in vitro tissue performance in applications where critical quality attributes must be quantified, like in drug development and screening.

3.
Arthritis Rheum ; 64(6): 1940-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22231515

RESUMO

OBJECTIVE: Mutations in LMNA encoding the A-type lamins cause several diseases, including those with features of premature aging and skeletal abnormalities. The aim of this study was to examine the expression of lamin A in cartilage from patients with osteoarthritis (OA) and the effects of its overexpression on chondrocyte senescence and apoptosis. METHODS: Human chondrocyte-like cells (SW-1353) were used. RNA isolated from human OA and non-OA cartilage was used for profiling messenger RNA expression, using Affymetrix microarray analysis. The effects of lamin A overexpression on mitochondrial function and apoptosis were examined by assessing mitochondrial membrane potential, ATP levels, and cytochrome c release, and with a TUNEL assay. Western blotting was performed to determine protein expression. RESULTS: Lamin A expression was markedly elevated in OA cartilage samples compared with non-OA control samples. Western blot analysis confirmed increased expression of lamin A in OA compared with non-OA cartilage. Interleukin-1ß treatment inhibited lamin A accumulation, whereas treatment with prostaglandin E(2) (PGE(2) ) caused a marked increase in lamin A accumulation. These effects of exogenous PGE(2) on lamin A expression were mediated via the EP(2) /EP(4) receptors. Transfected chondrocytes that expressed lamin A displayed markers of early senescence/apoptosis. CONCLUSION: The results of this study suggest that lamin A is up-regulated in OA chondrocytes, and that increased nuclear accumulation of lamin A in response to catabolic stress may account for the premature aging phenotype and apoptosis of OA chondrocytes.


Assuntos
Apoptose/fisiologia , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Lamina Tipo A/metabolismo , Osteoartrite do Joelho/metabolismo , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Cartilagem Articular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Dinoprostona/farmacologia , Feminino , Humanos , Interleucina-1beta/farmacologia , Lamina Tipo A/genética , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/genética
4.
PLoS One ; 18(1): e0278844, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36701370

RESUMO

Leiomyosarcoma (LMS) is an aggressive, often poorly differentiated cancer of the smooth muscle (SM) lineage for which the molecular drivers of transformation and progression are poorly understood. In microRNA (miRNA) profiling studies, miR-130b was previously found to be upregulated in LMS vs. normal SM, and down-regulated during the differentiation of mesenchymal stem cells (MSCs) into SM, suggesting a role in LMS tumor progression. In the present study, the effects of miR-130b on human LMS tumorigenesis were investigated. Stable miR-130b overexpression enhanced invasion of LMS cells in vitro, and led to the formation of undifferentiated, pleomorphic tumors in vivo, with increased growth and metastatic potential compared to control LMS cells. TSC1 was identified as a direct miR-130b target in luciferase-3'UTR assays, and shRNA-mediated knockdown of TSC1 replicated miR-130b effects. Loss-of-function and gain-of-function studies showed that miR-130b levels regulate cell morphology and motility. Following miR-130b suppression, LMS cells adopted a rounded morphology, amoeboid mode of cell movement and enhanced invasive capacity that was Rho/ROCK dependent. Conversely, miR-130b-overexpressing LMS cells exhibited Rho-independent invasion, accompanied by down-regulation of Rho-pathway effectors. In mesenchymal stem cells, both miR-130b overexpression and TSC1 silencing independently impaired SM differentiation in vitro. Together, the data reveal miR-130b as a pro-oncogenic miRNA in LMS and support a miR-130b-TSC1 regulatory network that enhances tumor progression via inhibition of SM differentiation.


Assuntos
Leiomiossarcoma , MicroRNAs , Humanos , Linhagem Celular Tumoral , Leiomiossarcoma/genética , MicroRNAs/genética , RNA Interferente Pequeno , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Proliferação de Células
5.
Front Bioeng Biotechnol ; 10: 901317, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35837555

RESUMO

In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body's healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.

6.
J Biol Chem ; 285(40): 31055-65, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20650891

RESUMO

Skeletogenesis depends on the activity of bone-forming cells derived from mesenchymal cells. The pathways that control mesenchymal cell differentiation are not well understood. We propose that Foxo1 is an early molecular regulator during mesenchymal cell differentiation into osteoblasts. In mouse embryos, Foxo1 expression is higher in skeletal tissues, while Foxo1 silencing has a drastic impact on skeletogenesis and craniofacial development, specially affecting pre-maxilla, nasal bone, mandible, tibia, and clavicle. Similarly, Foxo1 activity and expression increase in mouse mesenchymal cells under the influence of osteogenic stimulants. In addition, silencing Foxo1 blocks the expression of osteogenic markers such as Runx2, alkaline phosphatase, and osteocalcin and results in decreased culture calcification even in the presence of strong osteogenic stimulants. Conversely, the expression of these markers increases significantly in response to Foxo1 overexpression. One mechanism through which Foxo1 affects mesenchymal cell differentiation into osteoblasts is through regulation of a key osteogenic transcription factor, Runx2. Indeed, our results show that Foxo1 directly interacts with the promoter of Runx2 and regulates its expression. Using a tibia organ culture model, we confirmed that silencing Foxo1 decreases the expression of Runx2 and impairs bone formation. Furthermore, our data reveals that Runx2 and Foxo1 interact with each other and cooperate in the transcriptional regulation of osteoblast markers. In conclusion, our in vitro, ex vivo, and in vivo results strongly support the notion that Foxo1 is an early molecular regulator in the differentiation of mesenchymal cells into osteoblast.


Assuntos
Calcificação Fisiológica/fisiologia , Diferenciação Celular/fisiologia , Embrião de Mamíferos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Embrião de Mamíferos/citologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Inativação Gênica , Camundongos , Especificidade de Órgãos/fisiologia , Osteoblastos/citologia , Regiões Promotoras Genéticas/fisiologia
7.
FASEB J ; 23(1): 79-89, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18780763

RESUMO

In osteoarthritis (OA) articular chondrocytes undergo phenotypic changes culminating in the progressive loss of cartilage from the joint surface. The molecular mechanisms underlying these changes are poorly understood. Here we report enhanced (approximately 7-fold) expression of F-spondin, a neuronal extracellular matrix glycoprotein, in human OA cartilage (P<0.005). OA-specific up-regulation of F-spondin was also demonstrated in rat knee cartilage following surgical menisectomy. F-spondin treatment of OA cartilage explants caused a 2-fold increase in levels of the active form of TGF-beta1 (P<0.01) and a 10-fold induction of PGE2 (P<0.005) in culture supernatants. PGE2 induction was found to be dependent on TGF-beta and the thrombospondin domain of the F-spondin molecule. F-spondin addition to cartilage explant cultures also caused a 4-fold increase in collagen degradation (P<0.05) and a modest reduction in proteoglycan synthesis (approximately 20%; P<0.05), which were both TGF-beta and PGE2 dependent. F-spondin treatment also led to increased secretion and activation of MMP-13 (P<0.05). Together these studies identify F-spondin as a novel protein in OA cartilage, where it may act in situ at lesional areas to activate latent TGF-beta and induce cartilage degradation via pathways that involve production of PGE2.


Assuntos
Cartilagem Articular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Osteoartrite/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/metabolismo , Proteínas da Matriz Extracelular/genética , Humanos , Ratos , Ratos Sprague-Dawley , Regulação para Cima
8.
J Immunol ; 181(7): 5082-8, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802112

RESUMO

Elevated levels of PGE(2) have been reported in synovial fluid and cartilage from patients with osteoarthritis (OA). However, the functions of PGE(2) in cartilage metabolism have not previously been studied in detail. To do so, we cultured cartilage explants, obtained from patients undergoing knee replacement surgery for advanced OA, with PGE(2) (0.1-10 muM). PGE(2) inhibited proteoglycan synthesis in a dose-dependent manner (maximum 25% inhibition (p < 0.01)). PGE(2) also induced collagen degradation, in a manner inhibitable by the matrix metalloproteinase (MMP) inhibitor ilomastat. PGE(2) inhibited spontaneous MMP-1, but augmented MMP-13 secretion by OA cartilage explant cultures. PCR analysis of OA chondrocytes treated with PGE(2) with or without IL-1 revealed that IL-1-induced MMP-13 expression was augmented by PGE(2) and significantly inhibited by the cycolooygenase 2 selective inhibitor celecoxib. Conversely, MMP-1 expression was inhibited by PGE(2), while celecoxib enhanced both spontaneous and IL-1-induced expression. IL-1 induction of aggrecanase 5 (ADAMTS-5), but not ADAMTS-4, was also enhanced by PGE(2) (10 muM) and reversed by celecoxib (2 muM). Quantitative PCR screening of nondiseased and end-stage human knee OA articular cartilage specimens revealed that the PGE(2) receptor EP4 was up-regulated in OA cartilage. Moreover, blocking the EP4 receptor (EP4 antagonist, AH23848) mimicked celecoxib by inhibiting MMP-13, ADAMST-5 expression, and proteoglycan degradation. These results suggest that PGE(2) inhibits proteoglycan synthesis and stimulates matrix degradation in OA chondrocytes via the EP4 receptor. Targeting EP4, rather than cyclooxygenase 2, could represent a future strategy for OA disease modification.


Assuntos
Cartilagem Articular/metabolismo , Dinoprostona/fisiologia , Osteoartrite/metabolismo , Receptores de Prostaglandina E/fisiologia , Transdução de Sinais/imunologia , Idoso , Cartilagem Articular/enzimologia , Cartilagem Articular/patologia , Linhagem Celular , Células Cultivadas , Condrócitos/enzimologia , Condrócitos/metabolismo , Condrócitos/patologia , Dinoprostona/metabolismo , Ativação Enzimática/imunologia , Humanos , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Pessoa de Meia-Idade , Osteoartrite/enzimologia , Osteoartrite/patologia , Receptores de Prostaglandina E/biossíntese , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Técnicas de Cultura de Tecidos
9.
Cells ; 9(9)2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32927859

RESUMO

TSC1 is a tumor suppressor that inhibits cell growth via negative regulation of the mammalian target of rapamycin complex (mTORC1). TSC1 mutations are associated with Tuberous Sclerosis Complex (TSC), characterized by multiple benign tumors of mesenchymal and epithelial origin. TSC1 modulates self-renewal and differentiation in hematopoietic stem cells; however, its effects on mesenchymal stem cells (MSCs) are unknown. We investigated the impact of Tsc1 inactivation in murine bone marrow (BM)-MSCs, using tissue-specific, transgelin (Tagln)-mediated cre-recombination, targeting both BM-MSCs and smooth muscle cells. Tsc1 mutants were viable, but homozygous inactivation led to a dwarfed appearance with TSC-like pathologies in multiple organs and reduced survival. In young (28 day old) mice, Tsc1 deficiency-induced significant cell expansion of non-hematopoietic BM in vivo, and MSC colony-forming potential in vitro, that was normalized upon treatment with the mTOR inhibitor, everolimus. The hyperproliferative BM-MSC phenotype was lost in aged (1.5 yr) mice, and Tsc1 inactivation was also accompanied by elevated ROS and increased senescence. ShRNA-mediated knockdown of Tsc1 in BM-MSCs replicated the hyperproliferative BM-MSC phenotype and led to impaired adipogenic and myogenic differentiation. Our data show that Tsc1 is a negative regulator of BM-MSC proliferation and support a pivotal role for the Tsc1-mTOR axis in the maintenance of the mesenchymal progenitor pool.


Assuntos
Proliferação de Células , Células-Tronco Mesenquimais/citologia , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Esclerose Tuberosa/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Senescência Celular , Feminino , Camundongos , Camundongos Knockout , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/patologia
10.
Arthritis Rheum ; 58(9): 2786-97, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18759268

RESUMO

OBJECTIVE: To determine the effects of the antioxidant resveratrol on the functions of human chondrocytes in osteoarthritis (OA). METHODS: Chondrocytes and cartilage explants were isolated from OA patients undergoing knee replacement surgery. Effects of resveratrol in the presence or absence of interleukin-1beta (IL-1beta) stimulation were assessed by measurement of prostaglandin E(2) (PGE(2)) and leukotriene B(4) (LTB(4)) synthesis, cyclooxygenase (COX) activity, matrix metalloproteinase (MMP) expression, and proteoglycan production. To explore the mechanisms of action of resveratrol, its effects on mitochondrial function and apoptosis were examined by assessing mitochondrial membrane potential, ATP levels, cytochrome c release, and annexin V staining. RESULTS: Resveratrol inhibited both spontaneous and IL-1beta-induced PGE(2) production by >20% (P < 0.05) and by 80% (P < 0.001), respectively; similarly, LTB(4) production was reduced by >50% (P < 0.05). The production of PGE(2) was inhibited via a 70-90% suppression of COX-2 expression and enzyme activity (P < 0.05). Resveratrol also promoted anabolic effects in OA explant cultures, by elevating proteoglycan synthesis and decreasing production of MMPs 1, 3, and 13. Pretreatment of OA chondrocytes with resveratrol blocked mitochondrial membrane depolarization, loss of mitochondrial biomass, and IL-1beta-induced ATP depletion. Similarly, IL-1beta-mediated induction of the apoptotic markers cytochrome c and annexin V was also inhibited by resveratrol. Exogenous addition of PGE(2) abolished the protective effects of resveratrol on mitochondrial membrane integrity, ATP levels, expression of apoptotic markers, and DNA fragmentation. CONCLUSION: Resveratrol protects against IL-1beta-induced catabolic effects and prevents chondrocyte apoptosis via its inhibition of mitochondrial membrane depolarization and ATP depletion. These beneficial effects of resveratrol are due, in part, to its capacity to inhibit COX-2-derived PGE(2) synthesis. Resveratrol may therefore protect against oxidant injury and apoptosis, which are main features of progressive OA.


Assuntos
Trifosfato de Adenosina/metabolismo , Apoptose/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Osteoartrite/metabolismo , Estilbenos/farmacologia , Análise de Variância , Anexina A5/metabolismo , Antioxidantes/farmacologia , Western Blotting , Cartilagem/metabolismo , Condrócitos/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Dinoprostona/biossíntese , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-1beta/farmacologia , Leucotrieno B4/biossíntese , Metaloproteinases da Matriz/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Osteoartrite/tratamento farmacológico , Proteoglicanas/biossíntese , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Biomaterials ; 29(7): 904-16, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18045683

RESUMO

Ruptures of the anterior cruciate ligament (ACL) are common knee injuries that do not heal, even with surgical repair. Our research is directed towards developing novel, biological approaches that enable suture repair of this ligament. One promising strategy involves the insertion of a collagen hydrogel between the severed ends of the ACL. Cells migrate from the damaged ligament into the hydrogel and produce repair tissue. Here we have investigated the potential for augmenting this process by the transfer of insulin like growth factor (IGF) 1 cDNA to the repair cells using an adenovirus vector. The goal is to achieve direct, in situ gene delivery by loading the hydrogel with vector prior to its insertion into the defect. In a step-wise approach towards evaluating this process, we confirmed that monolayers of ACL fibroblasts were efficiently transduced by adenovirus vectors and continued to express transgenes when subsequently incorporated into the hydrogel; indeed, transgene expression persisted longer within collagen gels than in monolayer culture. Transfer of IGF-1 cDNA increased the cellularity of the gels and led to the synthesis and deposition of increased amounts of types I and III collagen, elastin, tenascin, and vimentin. The cells remained viable, even when subjected to high viral loads. Similar results were obtained when collagen hydrogels were preloaded with adenovirus prior to insertion into an experimental ACL lesion in vitro. These data confirm the promise of using vector-laden hydrogels for the in situ delivery of genes to cells within damaged ligaments and suggest novel possibilities for the biological repair of the ACL.


Assuntos
Lesões do Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Transgenes/genética , Adenoviridae/genética , Ligamento Cruzado Anterior/citologia , Biomarcadores , Sobrevivência Celular , Células Cultivadas , Colágeno , Fibroblastos , Regulação da Expressão Gênica , Humanos , Hidrogéis , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/genética
12.
Tissue Eng ; 13(9): 2227-37, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17561802

RESUMO

To investigate the use of a scaffold seeded with genetically modified meniscal cells or mesenchymal stem cells (MSCs) for the healing of meniscal lesions, primary meniscus cells and bone marrow-derived MSCs were isolated from bovine calves and transduced with first-generation adenoviral vectors encoding green fluorescent protein, luciferase, or transforming growth factor (TGF)-beta1 complementary deoxyribonucleic acid (cDNA). The genetically modified cells were seeded in type I collagen-glycosaminoglycan (GAG) matrices and transplanted into tears of the avascular zone of bovine menisci. After 3 weeks of in vitro culture, constructs and repair tissues were analyzed histologically, biochemically, and using reverse transcriptase polymerase chain reaction. Recombinant adenovirus readily transduced meniscal cells and MSCs, and transgene expression remained high after the cells were incorporated into collagen-GAG matrices. Transfer of TGF-beta1 cDNA increased cellularitiy and the synthesis of GAG/DNA [microg/microg]. It also led to stronger staining for proteoglycans and type II collagen and enhanced expression of meniscal genes. Transplantation of the TGF-beta1 transduced constructs into meniscal lesions of the avascular zone resulted in filling of the lesions with repair tissue after 3 weeks of in vitro culture. These results indicate that TGF-beta1 cDNA delivery may affect cell-based meniscus repair approaches in vivo.


Assuntos
DNA Complementar , Técnicas de Transferência de Genes , Engenharia Genética , Meniscos Tibiais , Engenharia Tecidual , Fator de Crescimento Transformador beta1/genética , Animais , Bovinos , Células Cultivadas , Genes Reporter , Humanos , Meniscos Tibiais/citologia , Células-Tronco Mesenquimais , Proteínas Recombinantes/genética
13.
Tissue Eng ; 13(8): 1987-93, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17518747

RESUMO

Facilitated endogenous repair is a novel approach to tissue engineering that avoids the ex vivo culture of autologous cells and the need for manufactured scaffolds, while minimizing the number and invasiveness of associated clinical procedures. The strategy relies on harnessing the intrinsic regenerative potential of endogenous tissues using molecular stimuli, such as gene transfer, to initiate reparative processes in situ. In the simplest example, direct percutaneous injection of an osteogenic vector is used to stimulate bone healing. If necessary, additional progenitor cells and space-filling scaffolds can be provided by autologous bone marrow, muscle, fat, and perhaps other tissues. These can be harvested, processed, and reimplanted by simple, expedited, intraoperative procedures. Examples of repair of experimental osseous and osteochondral lesions in laboratory animals are described. If successful, these strategies will provide methods for tissue regeneration that are not only effective but also inexpensive, safe, and clinically expeditious. Although orthopaedic examples are given here, the technology should be more generally applicable.


Assuntos
Engenharia Tecidual/economia , Engenharia Tecidual/métodos , Cicatrização/fisiologia , Animais , Humanos , Engenharia Tecidual/tendências
14.
ACS Biomater Sci Eng ; 2(10): 1787-1795, 2016 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-33440476

RESUMO

The demands of tissue engineering have driven a tremendous amount of research effort in 3D tissue culture technology and, more recently, in 3D printing. The need to use 3D tissue culture techniques more broadly in all of cell biology is well-recognized, but the transition to 3D has been impeded by the convenience, effectiveness, and ubiquity of 2D culture materials, assays, and protocols, as well as the lack of 3D counterparts of these tools. Interestingly, progress and discoveries in 3D bioprinting research may provide the technical support needed to grow the practice of 3D culture. Here we investigate an integrated approach for 3D printing multicellular structures while using the same platform for 3D cell culture, experimentation, and assay development. We employ a liquid-like solid (LLS) material made from packed granular-scale microgels, which locally and temporarily fluidizes under the focused application of stress and spontaneously solidifies after the applied stress is removed. These rheological properties enable 3D printing of multicellular structures as well as the growth and expansion of cellular structures or dispersed cells. The transport properties of LLS allow molecular diffusion for the delivery of nutrients or small molecules for fluorescence-based assays. Here, we measure viability of 11 different cell types in the LLS medium, we 3D print numerous structures using several of these cell types, and we explore the transport properties in molecular time-release assays.

15.
Crit Rev Eukaryot Gene Expr ; 12(4): 259-73, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12641395

RESUMO

Articular cartilage is particularly vulnerable to injury and degenerative conditions, and has a limited capacity for self-repair. Although current clinical procedures cannot restore a normal articular surface, there are a growing number of proteins that may be used to augment a repair process, or protect cartilage from degeneration. Because proteins are often difficult to administer effectively, gene therapy approaches are being developed to provide their sustained synthesis at sites of injury or disease. To promote cartilage repair, cDNAs can be targeted to synovium, or cartilage. Gene transfer to the synovium is generally considered more suitable for chondroprotective therapies that rely on expression of large amounts of anti-inflammatory mediators. The delivery of genes to cartilage defects to promote enhanced repair can be performed by either direct administration of gene delivery vectors, or by implantation of genetically modified chondrogenic cells. Variations of these methods have been used to demonstrate that exogenous cDNAs encoding growth factors can be delivered locally to sites of cartilage damage where they are expressed at physiologically relevant levels. Data is beginning to emerge that suggests that delivery and expression of these genescan influence a repair response toward the synthesis of normal articular cartilage in vivo. This article reviews the current status of gene delivery for cartilage healing and presents some of the remaining challenges.


Assuntos
Cartilagem Articular/fisiologia , Terapia Genética , Regeneração/genética , Animais , Cartilagem Articular/cirurgia , Técnicas de Transferência de Genes , Humanos , Proteínas Recombinantes/genética
16.
Methods Mol Med ; 100: 147-64, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15280594

RESUMO

The major requirement of a successful gene transfer is the efficient delivery of an exogenous therapeutic gene to the appropriate cell type with subsequent high or regulated levels of expression. In this context, viral systems are more efficient than nonviral systems, giving higher levels of gene expression for longer periods. For the application of osteoarthritis (OA), gene products triggering anti-inflammatory or chondroprotective effects are of obvious therapeutic utility. Thus, their cognate genes are candidates for use in the gene therapy of OA. In this chapter, we describe the preparation, the use, and the effect of the transduction of chondrocytes or synovial fibroblasts with an adenoviral vector encoding the cDNA for glutamine: fructose-6-phosphate amidotransferase (GFAT). This is intended to serve as an example of a technology that can be used to evaluate the biological effects of overexpression of other cDNAs.


Assuntos
Adenoviridae/genética , Condrócitos/metabolismo , Fibroblastos/metabolismo , Vetores Genéticos/genética , Membrana Sinovial/citologia , Transdução Genética/métodos , Adenoviridae/química , Cartilagem Articular/química , Cartilagem Articular/citologia , Cartilagem Articular/metabolismo , Condrócitos/química , DNA Recombinante/química , DNA Recombinante/genética , Fibroblastos/química , Terapia Genética/métodos , Vetores Genéticos/química , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/genética , Humanos , Interleucina-1/farmacologia , Óxido Nítrico/biossíntese , Osteoartrite/genética , Osteoartrite/terapia , Plasmídeos/química , Plasmídeos/genética
17.
Biorheology ; 40(1-3): 61-72, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12454388

RESUMO

The effects of hypotonic (180 mOsm) and hypertonic (580 mOsm) medium loading on chondrocyte aggrecan gene expression in 2D monolayer and 3D hydrogel culture (agarose or alginate) were studied. Aggrecan promoter activity was monitored using a luciferase reporter gene assay and transient transfection. Osmotic loading was observed to differentially affect promoter activity, with hypotonic loading generally producing at least a 40% elevation in promoter activity, except for the case of alginate where a 50% suppression was observed. Hypertonic loading produced at least a 35% decrease in activity for all cultures. Similar osmolality-induced changes to aggrecan mRNA levels were observed in monolayer cells using qPCR. Deletion of exon 1 blocked the sensitivity of monolayer cells to hypertonic but not hypotonic medium changes. Confocal microscopy measurements suggested that the degree of hypotonic swelling in cells encapsulated in 3D matrix was restricted compared to monolayer cells whereas the degree of hypertonic shrinking was similar under both culture conditions.


Assuntos
Cartilagem Articular/citologia , Condrócitos/metabolismo , Proteínas da Matriz Extracelular , Proteoglicanas/biossíntese , Agrecanas , Animais , Bovinos , Técnicas de Cultura de Células , Tamanho Celular/fisiologia , Regulação da Expressão Gênica , Soluções Hipertônicas , Soluções Hipotônicas , Lectinas Tipo C , Concentração Osmolar , Proteoglicanas/genética , RNA Mensageiro/genética
18.
PLoS One ; 9(5): e98388, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24875054

RESUMO

F-spondin is a pericellular matrix protein upregulated in developing growth plate cartilage and articular cartilage during osteoarthritis. To address its function in bone and cartilage in vivo, we generated mice that were deficient for the F-spondin gene, Spon1. Spon1-/- mice were viable and developed normally to adulthood with no major skeletal abnormalities. At 6 months, femurs and tibiae of Spon1-/- mice exhibited increased bone mass, evidenced by histological staining and micro CT analyses, which persisted up to 12 months. In contrast, no major abnormalities were observed in articular cartilage at any age group. Immunohistochemical staining of femurs and tibiae revealed increased levels of periostin, alkaline phosphate and tartrate resistant acid phosphatase (TRAP) activity in the growth plate region of Spon1-/- mice, suggesting elevated bone synthesis and turnover. However, there were no differences in serum levels of TRAP, the bone resorption marker, CTX-1, or osteoclast differentiation potential between genotypes. Knockout mice also exhibited reduced levels of TGF-ß1 in serum and cultured costal chondrocytes relative to wild type. This was accompanied by increased levels of the BMP-regulatory SMADs, P-SMAD1/5 in tibiae and chondrocytes. Our findings indicate a previously unrecognized role for Spon1 as a negative regulator of bone mass. We speculate that Spon1 deletion leads to a local and systemic reduction of TGF-ß levels resulting in increased BMP signaling and increased bone deposition in adult mice.


Assuntos
Osso e Ossos/anatomia & histologia , Osso e Ossos/metabolismo , Proteínas da Matriz Extracelular/deficiência , Fenótipo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Osso e Ossos/citologia , Osso e Ossos/diagnóstico por imagem , Condrócitos/metabolismo , Ordem dos Genes , Marcação de Genes , Genótipo , Camundongos , Camundongos Knockout , Osteoclastos/citologia , Osteoclastos/metabolismo , Fosforilação , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microtomografia por Raio-X
19.
J Orthop Res ; 28(10): 1323-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20839318

RESUMO

This study examines the role of F-spondin, an extracellular matrix protein of osteoarthritic cartilage, during chondrocyte maturation in embryonic growth plate cartilage. In chick tibia, F-spondin expression localized to the hypertrophic and calcified zones of the growth plate. Functional studies using tibial organ cultures indicated that F-spondin inhibited (∼35%, p = 0.02), and antibodies to F-spondin increased (∼30%, p < 0.1) longitudinal limb growth relative to untreated controls. In cell cultures, induction of chondrocyte maturation, by retinoic acid (RA) or transforming growth factor (TGF)-ß treatment led to a significant upregulation of F-spondin (p < 0.05). F-spondin transfection increased mineral deposition, alkaline phosphatase (AP) and matrix metalloproteinase (MMP)-13 mRNA levels (p < 0.05), and AP activity following RA stimulation, compared to mock transfected controls. Using AP as a differentiation marker we then investigated the mechanism of F-spondin promaturation effects. Blocking endogenous F-spondin via its thrombospondin (TSR) domain inhibited RA induced AP activity 40% compared to controls (p < 0.05). The stimulatory effect of F-spondin on AP expression was also inhibited following depletion of TGF-ß from culture supernatants. Our findings indicate that F-spondin is expressed in embryonic cartilage, where it has the capacity to enhance chondrocyte terminal differentiation and mineralization via interactions in its TSR domain and TGF-ß dependent pathways.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/citologia , Proteínas da Matriz Extracelular/fisiologia , Osteogênese/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Feminino , Lâmina de Crescimento/citologia , Lâmina de Crescimento/fisiologia , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos , Modelos Animais , Gravidez , Fator de Crescimento Transformador beta/farmacologia , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA