Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 31(2): 420-434, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36245128

RESUMO

An estimated 100,000 patients each year in the United States suffer severe disability from bone defects that fail to heal, a condition where bone-regenerative therapies could provide substantial clinical benefits. Although recombinant human bone morphogenetic protein-2 (rhBMP2) is an osteogenic growth factor that is clinically approved for this purpose, it is only effective when used at exceedingly high doses that incur substantial costs, induce severe inflammation, produce adverse side effects, and form morphologically abnormal bone. Using a validated rat femoral segmental defect model, we show that bone formed in response to clinically relevant doses of rhBMP2 is accompanied by elevated expression of interleukin-1 (IL-1). Local delivery of cDNA encoding the IL-1 receptor antagonist (IL-1Ra) achieved bridging of segmental, critical size defects in bone with a 90% lower dose of rhBMP2. Unlike use of high-dose rhBMP2, bone formation in the presence of IL-1Ra occurred via the native process of endochondral ossification, resulting in improved quality without sacrificing the mechanical properties of the regenerated bone. Our results demonstrate that local immunomodulation may permit effective use of growth factors at lower doses to recapitulate more precisely the native biology of healing, leading to higher-quality tissue regeneration.


Assuntos
Proteína Antagonista do Receptor de Interleucina 1 , Osteogênese , Humanos , Ratos , Animais , Osteogênese/genética , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Regeneração Óssea/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/farmacologia
2.
Mol Ther ; 26(1): 208-218, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29107477

RESUMO

Because muscle contains osteoprogenitor cells and has a propensity to form bone, we have explored its utility in healing large osseous defects. Healing is achieved by the insertion of muscle fragments transduced with adenovirus encoding BMP-2 (Ad.BMP-2). However, it is not known whether the genetically modified muscle contributes osteoprogenitor cells to healing defects or merely serves as a local source of BMP-2. This question is part of the larger debate on the fate of progenitor cells introduced into sites of tissue damage to promote regeneration. To address this issue, we harvested fragments of muscle from rats constitutively expressing GFP, transduced them with Ad.BMP-2, and implanted them into femoral defects in wild-type rats under various conditions. GFP+ cells persisted within defects for the entire 8 weeks of the experiments. In the absence of bone formation, these cells presented as fibroblasts. When bone was formed, GFP+ cells were present as osteoblasts and osteocytes and also among the lining cells of new blood vessels. The genetically modified muscle thus contributed progenitor cells as well as BMP-2 to the healing defect, a property of great significance in light of the extensive damage to soft tissue and consequent loss of endogenous progenitors in problematic fractures.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Osteoblastos/metabolismo , Osteogênese , Absorciometria de Fóton , Animais , Biópsia , Regeneração Óssea , Expressão Gênica , Genes Reporter , Imuno-Histoquímica , Masculino , Músculo Esquelético/metabolismo , RNA Mensageiro/genética , Ratos , Cicatrização , Microtomografia por Raio-X
3.
J Biol Chem ; 288(41): 29494-505, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23970554

RESUMO

Bone marrow contains mesenchymal stem cells (MSCs) that can differentiate along multiple mesenchymal lineages. In this capacity they are thought to be important in the intrinsic turnover and repair of connective tissues while also serving as a basis for tissue engineering and regenerative medicine. However, little is known of the biological responses of human MSCs to inflammatory conditions. When cultured with IL-1ß, marrow-derived MSCs from 8 of 10 human subjects deposited copious hydroxyapatite, in which authenticity was confirmed by Fourier transform infrared spectroscopy. Transmission electron microscopy revealed the production of fine needles of hydroxyapatite in conjunction with matrix vesicles. Alkaline phosphatase activity did not increase in response to inflammatory mediators, but PPi production fell, reflecting lower ectonucleotide pyrophosphatase activity in cells and matrix vesicles. Because PPi is the major physiological inhibitor of mineralization, its decline generated permissive conditions for hydroxyapatite formation. This is in contrast to MSCs treated with dexamethasone, where PPi levels did not fall and mineralization was fuelled by a large and rapid increase in alkaline phosphatase activity. Bone sialoprotein was the only osteoblast marker strongly induced by IL-1ß; thus these cells do not become osteoblasts despite depositing abundant mineral. RT-PCR did not detect transcripts indicative of alternative mesenchymal lineages, including chondrocytes, myoblasts, adipocytes, ligament, tendon, or vascular smooth muscle cells. IL-1ß phosphorylated multiple MAPKs and activated nuclear factor-κB (NF-κB). Certain inhibitors of MAPK and PI3K, but not NF-κB, prevented mineralization. The findings are of importance to soft tissue mineralization, tissue engineering, and regenerative medicine.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Citocinas/farmacologia , Durapatita/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Adulto , Idoso , Idoso de 80 Anos ou mais , Fosfatase Alcalina/metabolismo , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/ultraestrutura , Cálcio/metabolismo , Células Cultivadas , Difosfatos/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Sialoproteína de Ligação à Integrina/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Fenótipo , Diester Fosfórico Hidrolases/metabolismo , Fosforilação/efeitos dos fármacos , Pirofosfatases/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
4.
Biochimie ; 207: 33-48, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36427681

RESUMO

Mesenchymal stem/stromal cells (MSCs) are multipotent somatic cells that have been widely explored in the field of regenerative medicine. MSCs possess the ability to secrete soluble factors as well as lipid bound extracellular vesicles (EVs). MSCs have gained increased interest and attention as a result of their therapeutic properties, which are thought to be attributed to their secretome. However, while the use of MSCs as whole cells pose heterogeneity concerns and survival issues post-transplantation, such limitations are absent in cell-free EV-based treatments. EVs derived from MSCs are promising therapeutic agents for a range of clinical conditions and disorders owing to their immunomodulatory, pro-regenerative, anti-inflammatory, and antifibrotic activity. Recent successes with preclinical studies using EVs for repair and regeneration of damaged tissues such as cardiac tissue, lung, liver, pancreas, bone, skin, cornea, and blood diseases are discussed in this review. We also discuss delivery strategies of EVs using biomaterials as delivery vehicles through systemic or local administration. Despite its effectiveness in preclinical investigations, the application of MSC-EV in clinical settings will necessitate careful consideration surrounding issues such as: i) scalability and isolation, ii) biodistribution, iii) targeting specific tissues, iv) quantification and characterization, and v) safety and efficacy of dosage. The future of EVs in regenerative medicine is promising yet still needs further investigation on enhancing the efficacy, scalability, and potency for clinical applications.


Assuntos
Vesículas Extracelulares , Mesoderma , Regeneração , Medicina Regenerativa , Células-Tronco , Vesículas Extracelulares/classificação , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Medicina Regenerativa/métodos , Medicina Regenerativa/normas , Medicina Regenerativa/tendências , Mesoderma/citologia , Células-Tronco/citologia , Humanos , Animais , Biotecnologia/métodos , Biotecnologia/normas , Biotecnologia/tendências
5.
Nat Commun ; 14(1): 3616, 2023 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-37330524

RESUMO

NAD is an essential co-factor for cellular energy metabolism and multiple other processes. Systemic NAD+ deficiency has been implicated in skeletal deformities during development in both humans and mice. NAD levels are maintained by multiple synthetic pathways but which ones are important in bone forming cells is unknown. Here, we generate mice with deletion of Nicotinamide Phosphoribosyltransferase (Nampt), a critical enzyme in the NAD salvage pathway, in all mesenchymal lineage cells of the limbs. At birth, NamptΔPrx1 exhibit dramatic limb shortening due to death of growth plate chondrocytes. Administration of the NAD precursor nicotinamide riboside during pregnancy prevents the majority of in utero defects. Depletion of NAD post-birth also promotes chondrocyte death, preventing further endochondral ossification and joint development. In contrast, osteoblast formation still occurs in knockout mice, in line with distinctly different microenvironments and reliance on redox reactions between chondrocytes and osteoblasts. These findings define a critical role for cell-autonomous NAD homeostasis during endochondral bone formation.


Assuntos
Metabolismo Energético , NAD , Humanos , Camundongos , Animais , NAD/metabolismo , Oxirredução , Homeostase , Camundongos Knockout , Citocinas/metabolismo
6.
bioRxiv ; 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37790307

RESUMO

Multivalency enables nanostructures to bind molecular targets with high affinity. Although antibodies can be generated against a wide range of antigens, their shape and size cannot be tuned to match a given target. DNA nanotechnology provides an attractive approach for designing customized multivalent scaffolds due to the addressability and programmability of the nanostructure shape and size. Here, we design a nanoscale synthetic antibody ("nano-synbody") based on a three-helix bundle DNA nanostructure with one, two, or three identical arms terminating in a mini-binder protein that targets the SARS-CoV-2 spike protein. The nano-synbody was designed to match the valence and distance between the three receptor binding domains (RBDs) in the spike trimer, in order to enhance affinity. The protein-DNA nano-synbody shows tight binding to the wild-type, Delta, and several Omicron variants of the SARS-CoV-2 spike trimer, with affinity increasing as the number of arms increases from one to three. The effectiveness of the nano-synbody was also verified using a pseudovirus neutralization assay, with the three-arm nanostructure inhibiting two Omicron variants against which the structures with only one or two arms are ineffective. The structure of the three-arm nano-synbody bound to the Omicron variant spike trimer was solved by negative-stain transmission electron microscopy reconstruction, and shows the protein-DNA nanostructure with all three arms attached to the RBD domains, confirming the intended trivalent attachment. The ability to tune the size and shape of the nano-synbody, as well as its potential ability to attach two or more different binding ligands, will enable the high-affinity targeting of a range of proteins not possible with traditional antibodies.

7.
Eur Cell Mater ; 23: 94-101; discussion 101-2, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-22354460

RESUMO

Tendon rupture is a common injury. Inadequate endogenous repair often leaves patients symptomatic, with tendons susceptible to re-rupture. Administration of certain growth factors improves tendon healing in animal models, but their delivery remains a challenge. Here we evaluated the delivery of TGF-ß1 to tendon defects by the implantation of genetically modified muscle grafts. Rat muscle biopsies were transduced with recombinant adenovirus encoding TGF-ß1 and grafted onto surgically transected Achilles tendons in recipient animals. Tissue regenerates were compared to those of controls by biomechanical testing as well as histochemical and immunohistochemical analyses. Healing was greatly accelerated when genetically modified grafts were implanted into tendon defects, with the resulting repair tissue gaining nearly normal histological appearance as early as 2 weeks postoperatively. This was associated with decreased deposition of type III collagen in favour of large fibre bundles indicative of type I collagen. These differences in tendon composition coincided with accelerated restoration of mechanical strength. Tendon thickness increased in gene-treated animals at weeks 1 and 2, but by week 8 became significantly lower than that of controls suggesting accelerated remodelling. Thus localised TGF-ß1 delivery via adenovirus-modified muscle grafts improved tendon healing in this rat model and holds promise for clinical application.


Assuntos
Tendão do Calcâneo/cirurgia , Terapia Genética , Músculo Esquelético/transplante , Traumatismos dos Tendões/cirurgia , Fator de Crescimento Transformador beta1/administração & dosagem , Fator de Crescimento Transformador beta1/genética , Adenoviridae , Animais , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , DNA Complementar , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Ruptura , Estresse Mecânico , Traumatismos dos Tendões/metabolismo , Transdução Genética , Cicatrização
8.
Cartilage ; 13(2_suppl): 1720S-1733S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34809478

RESUMO

OBJECTIVE: To support the preclinical evaluation of therapeutics that target chondrogenesis, our goal was to generate a rat strain that can noninvasively report endogenous chondrogenic activity. DESIGN: A transgene was constructed in which the dual expression of bioluminescent (firefly luciferase) and fluorescent (mCherry) reporters is controlled by regulatory sequences from rat Col2a1. Candidate lines were established on a Lewis background and characterized by serial bioluminescence imaging as well as ex vivo measurement of molecular reporter levels in several tissues. The sensitivity and specificity of the reporter strain were assessed in models of orthotopic and ectopic chondrogenesis. RESULTS: Substantial bioluminescence signal was detected from cartilaginous regions, including the appendicular synovial joints, spine, sternum, nose, and pinnae. Bioluminescent radiance was intense at 1 month of age, rapidly declined with continued development, yet remained detectable in 2-year-old animals. Explant imaging and immunohistochemistry confirmed that both molecular reporters were localized to cartilage. Implantation of wild-type bone marrow stromal cells into osteochondral defects made in both young adult and aged reporter rats led to a time-dependent elevation of intra-articular reporter activity concurrent with cartilaginous tissue repair. To stimulate ectopic, endochondral bone formation, bone morphogenetic protein 2 was overexpressed in the gastrocnemius muscle, which led to bioluminescent signal that closely preceded heterotopic ossification. CONCLUSIONS: This strain can help develop strategies to stimulate cartilage repair and endochondral bone formation or to inhibit chondrogenesis associated with heterotopic ossification.


Assuntos
Condrogênese , Engenharia Tecidual , Animais , Condrogênese/genética , Osteogênese , Ratos , Ratos Endogâmicos Lew , Ratos Transgênicos , Engenharia Tecidual/métodos
9.
J Control Release ; 318: 109-123, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31843642

RESUMO

Targeted drug delivery to joint tissues like cartilage remains a challenge that has prevented clinical translation of promising osteoarthritis (OA) drugs. Local intra-articular (IA) injections of drugs suffer from rapid clearance from the joint space and slow diffusive transport through the dense, avascular cartilage matrix comprised of negatively charged glycosaminoglycans (GAGs). Here we apply drug carriers that leverage electrostatic interactions with the tissue's high negative fixed charge density (FCD) for delivering small molecule drugs to cartilage cell and matrix sites. We demonstrate that a multi-arm cationic nano-construct of Avidin (mAv) with 28 sites for covalent drug conjugation can rapidly penetrate through the full thickness of cartilage in high concentration and have long intra-cartilage residence time in both healthy and arthritic cartilage via weak-reversible binding with negatively charged aggrecans. mAv's intra-cartilage mean uptake was found to be 112× and 33× the equilibration bath concentration in healthy and arthritic (50% GAG depleted) cartilage, respectively. mAv was conjugated with Dexamethasone (mAv-Dex), a broad-spectrum glucocorticoid, using a combination of hydrolysable ester linkers derived from succinic anhydride (SA), 3,3-dimethylglutaric anhydride (GA) and phthalic anhydride (PA) in 2:1:1 M ratio that enabled 50% drug release within 38.5 h followed by sustained release in therapeutic doses over 2 weeks. A single 10 µM low dose of controlled release mAv-Dex (2:1:1) effectively suppressed IL-1α-induced GAG loss, cell death and inflammatory response significantly better than unmodified Dex over 2 weeks in cartilage explant culture models of OA. With this multi-arm design, <1 µM Avidin was needed - a concentration which has been shown to be safe, preventing further GAG loss and cytotoxicity. A charge-based cartilage homing drug delivery platform like this can elicit disease modifying effects as well as facilitate long-term symptomatic pain and inflammation relief by enhancing tissue specificity and prolonging intra-cartilage residence time of OA drugs. This nano-construct thus has high translational potential for enabling intra-cartilage delivery of a broad array of small molecule OA drugs and their combinations to chondrocytes, enabling OA treatment with a single injection of low drug doses and eliminating toxicity issues associated with multiple high dose injections.


Assuntos
Cartilagem Articular , Osteoartrite , Avidina/uso terapêutico , Condrócitos , Portadores de Fármacos/uso terapêutico , Humanos , Injeções Intra-Articulares , Osteoartrite/tratamento farmacológico
10.
Aging Cell ; 19(11): e13247, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33048436

RESUMO

Marrow adipocytes and osteoblasts differentiate from common mesenchymal progenitors in a mutually exclusive manner, and diversion of these progenitors toward adipocytes in old age has been proposed to account for the decline in osteoblasts and the development of involutional osteoporosis. This idea has been supported by evidence that thiazolidinedione (TZD)-induced activation of PPARγ, the transcription factor required for adipocyte differentiation, increases marrow fat and causes bone loss. We functionally tested this hypothesis using C57BL/6J mice with conditional deletion of PPARγ from early mesenchymal progenitors targeted by the Prx1-Cre transgene. Using a longitudinal littermate-controlled study design, we observed that PPARγ is indispensable for TZD-induced increase in marrow adipocytes in 6-month-old male mice, and age-associated increase in marrow adipocytes in 22-month-old female mice. In contrast, PPARγ is dispensable for the loss of cortical and trabecular bone caused by TZD or old age. Instead, PPARγ restrains age-dependent development of cortical porosity. These findings do not support the long-standing hypothesis that increased marrow adipocyte differentiation contributes to bone loss in old age but reveal a novel role of mesenchymal cell PPARγ in the maintenance of cortical integrity.


Assuntos
Adipogenia/fisiologia , Osteoporose/fisiopatologia , Fatores Etários , Animais , Diferenciação Celular , Feminino , Camundongos
11.
Bone ; 121: 284-292, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30738214

RESUMO

The sirtuin family of NAD+-dependent protein deacetylases promotes longevity and counteracts age-related diseases. One of the major targets of Sirtuins are the FoxO family of transcription factors. FoxOs play a major role in the adaptation of cells to a variety of stressors such as oxidative stress and growth factor deprivation. Studies with murine models of cell-specific loss- or gain-of-function of Sirtuins or FoxOs and with Sirtuin1 stimulators have provided novel insights into the function and signaling of these proteins on the skeleton. These studies have revealed that both Sirtuins and FoxOs acting directly in cartilage and bone cells are critical for normal skeletal development, homeostasis and that their dysregulation might contribute to skeletal disease. Deacetylation of FoxOs by Sirt1 in osteoblasts and osteoclasts stimulates bone formation and inhibits bone resorption, making Sirt1 ligands promising therapeutic agents for diseases of low bone mass. While a similar link has not been established in chondrocytes, Sirt1 and FoxOs both have chondroprotective actions, suggesting that Sirt1 activators may have similar efficacy in preventing cartilage degeneration due to aging or injury. In this review we summarize these advances and discuss their implications for the pathogenesis of age-related osteoporosis and osteoarthritis.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Osteoporose/microbiologia , Sirtuínas/metabolismo , Animais , Autofagia/fisiologia , Senescência Celular/fisiologia , Condrócitos/metabolismo , Humanos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporose/metabolismo , Osteoporose/fisiopatologia
12.
Arthritis Res Ther ; 21(1): 238, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31722745

RESUMO

BACKGROUND: Most in vitro studies of potential osteoarthritis (OA) therapies have used cartilage monocultures, even though synovium is a key player in mediating joint inflammation and, thereby, cartilage degeneration. In the case of interleukin-1 (IL-1) inhibition using its receptor antagonist (IL-1Ra), like chondrocytes, synoviocytes also express IL-1 receptors that influence intra-articular IL-1 signaling and IL-1Ra efficacy. The short residence time of IL-1Ra after intra-articular injection requires the application of frequent dosing, which is clinically impractical and comes with increased risk of infection; these limitations motivate the development of effective drug delivery strategies that can maintain sustained intra-articular IL-1Ra concentrations with only a single injection. The goals of this study were to assess how the presence of synovium in IL-1-challenged cartilage-synovium co-culture impacts the time-dependent biological response of single and sustained doses of IL-1Ra, and to understand the mechanisms underlying any co-culture effects. METHODS: Bovine cartilage explants with or without synovium were treated with IL-1α followed by single or multiple doses of IL-1Ra. Effects of IL-1Ra in rescuing IL-1α-induced catabolism in cartilage monoculture and cartilage-synovium co-culture were assessed by measuring loss of glycosaminoglycans (GAGs) and collagen using DMMB (dimethyl-methylene blue) and hydroxyproline assays, respectively, nitric oxide (NO) release using Griess assay, cell viability by fluorescence staining, metabolic activity using Alamar blue, and proteoglycan biosynthesis by radiolabel incorporation. Day 2 conditioned media from mono and co-cultures were analyzed by mass spectrometry and cytokine array to identify proteins unique to co-culture that contribute to biological crosstalk. RESULTS: A single dose of IL-1Ra was ineffective, and a sustained dose was necessary to significantly suppress IL-1α-induced catabolism as observed by enhanced suppression of GAG and collagen loss, NO synthesis, rescue of chondrocyte metabolism, viability, and GAG biosynthesis rates. The synovium exhibited a protective role as the effects of single-dose IL-1Ra were significantly enhanced in cartilage-synovium co-culture and were accompanied by release of anti-catabolic factors IL-4, carbonic anhydrase-3, and matrilin-3. A total of 26 unique proteins were identified in conditioned media from co-cultures, while expression levels of many additional proteins important to cartilage homeostasis were altered in co-culture compared to monocultures; principal component analysis revealed distinct clustering between co-culture and cartilage and synovium monocultures, thereby confirming significant crosstalk. CONCLUSIONS: IL-1Ra suppresses cytokine-induced catabolism in cartilage more effectively in the presence of synovium, which was associated with endogenous production of anti-catabolic factors. Biological crosstalk between cartilage and synovium is significant; thus, their co-cultures should better model the intra-articular actions of potential OA therapeutics. Additionally, chondroprotective effects of IL-1Ra require sustained drug levels, underscoring the need for developing drug delivery strategies to enhance its joint residence time following a single intra-articular injection.


Assuntos
Cartilagem/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Citocinas/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/antagonistas & inibidores , Interleucina-1alfa/farmacologia , Membrana Sinovial/efeitos dos fármacos , Animais , Cartilagem/citologia , Cartilagem/metabolismo , Bovinos , Células Cultivadas , Condrócitos/metabolismo , Técnicas de Cocultura/métodos , Relação Dose-Resposta a Droga , Humanos , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Metabolismo/efeitos dos fármacos , Metabolismo/fisiologia , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo
13.
Tissue Eng Part C Methods ; 25(3): 176-190, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30727864

RESUMO

IMPACT STATEMENT: The promoter characterized in this study has been made accessible as a resource for the skeletal tissue engineering and regenerative medicine community. When combined with suitable reporter vectors, the resulting tools can be used for noninvasive and/or high-throughput screening of test conditions for stimulating chondrogenesis by candidate stem/progenitor cells. As demonstrated in this study, they can also be used with small animal imaging platforms to monitor the chondrogenic activity of implanted progenitors within orthotopic models of bone and cartilage repair.


Assuntos
Osso e Ossos/citologia , Condrócitos/citologia , Condrogênese , Articulações/fisiologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Engenharia Tecidual/métodos , Idoso , Animais , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Articulações/lesões , Masculino , Células-Tronco Mesenquimais/fisiologia , Ratos , Ratos Endogâmicos F344
14.
Tissue Eng ; 13(6): 1333-45, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17518712

RESUMO

Alginate hydrogel culture has been shown to reestablish chondrocytic phenotype following monolayer expansion; however, previous studies have not adequately addressed how culture conditions affect the signaling systems responsible for chondrocyte metabolic activity. Here we investigate whether chondrocyte culture history influences the insulin-like growth factor-I (IGF-I) signaling system and its regulation by interleukin-1 (IL-1). Articular chondrocytes (ACs) from equine stifle joints were expanded by serial passage and were either encapsulated in alginate beads or maintained in monolayer culture for 10 days. Alginate-derived cells (ADCs) and monolayer-derived cells (MDCs) were then plated at high density, stimulated with IL-1beta (1 and 10 ng/mL) or IGF-I (50 ng/mL) for 48 h, and assayed for levels of type I IGF receptor (IGF-IR), IGF binding proteins (IGFBPs), and endogenously secreted IGF-I. Intermediate alginate culture yielded relatively low IGF-IR levels that increased in response to IL-1beta, whereas higher receptor levels on MDCs were reduced by cytokine. MDCs also secreted substantially more IGFBP-2, the predominant binding protein in conditioned media (CM), though IL-1beta suppressed levels for both cell populations. Concentrations of autocrine/paracrine IGF-I paralleled IGFBP-2 secretion. Disparate basal levels of IGF-IR and IGFBP-2, but not IGF-I, were attributed to relative transcript expression. Systemic differences coincided with varied effects of IL-1beta and IGF-I on cell growth and type I collagen expression. We conclude that culture strategy impacts the IGF-I signaling system of ACs, potentially altering their capacity to mediate cartilage repair. Consideration of hormonal regulators may be an essential element to improve chondrocyte culture protocols used in tissue engineering applications.


Assuntos
Alginatos/química , Cartilagem Articular/fisiologia , Proliferação de Células/efeitos dos fármacos , Condrócitos/fisiologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Interleucina-1beta/administração & dosagem , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis/química , Cartilagem Articular/citologia , Cartilagem Articular/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Células Cultivadas , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Cavalos , Fator de Crescimento Insulin-Like I/metabolismo , Interleucina-1beta/metabolismo
15.
J Orthop Res ; 34(12): 2137-2145, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27037517

RESUMO

Orthopedic surgeons sometimes combine recombinant, human BMP-2 with autograft bone when dealing with problematic osseous fractures. Although some case reports indicate success with this off-label strategy, there have been no randomized controlled trials. Moreover, a literature search revealed only one pre-clinical study and this was in a cranial defect model. The present project examined the consequences of combining BMP-2 with particles of living bone in a rat femoral defect model. Human bone particles were recovered with a reamer-irrigator-aspirator (RIA). To allow acceptance of the xenograft as surrogate autograft, rats were administered an immunosuppressive cocktail that does not interfere with bone healing. Implantation of 200 µg living bone particles generated a small amount of new bone and defects did not heal. Graded amounts of BMP-2 that alone provoked no healing (1.1 µg), borderline healing (5.5 µg), or full healing (11 µg) were added to this amount of bone particles. Addition of BMP-2 (1.1 µg) increased osteogenesis, and produced bridging in 2 of 7 defects. The combination of BMP-2 (5.5 µg) and bone particles made healing more reliable and advanced the maturation of the regenerate. Bone formation with BMP-2 (11 µg) and bone particles showed improved maturation. Thus, the combination of autograft and BMP-2 may be helpful clinically under conditions where the healing response is suboptimal. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:2137-2145, 2016. Clinical significance These data support the clinical use of recombinant, human BMP-2 with autograft bone when treating large segmental osseous defects. The combination leads to greater bone formation and accelerates the maturation of the regenerate.


Assuntos
Proteína Morfogenética Óssea 2/administração & dosagem , Transplante Ósseo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Avaliação Pré-Clínica de Medicamentos , Fêmur , Humanos , Masculino , Pessoa de Meia-Idade , Distribuição Aleatória , Ratos Endogâmicos F344
16.
J Orthop Res ; 33(5): 660-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25627105

RESUMO

For evaluation of new approaches to drug delivery into cartilage, the choice of an animal model is critically important. Since cartilage thickness varies with animal size, different levels of drug uptake, transport and retention should be expected. Simple intra-articular injection can require very high drug doses to achieve a concentration gradient high enough for drug diffusion into cartilage. New approaches involve nanoparticle delivery of functionalized drugs directly into cartilage; however, diffusion-binding kinetics proceeds as the square of cartilage thickness. In this study, we demonstrate the necessity of using larger animals for sustained intra-cartilage delivery and retention, exemplified by intra-articular injection of Avidin (drug-carrier) into rabbits and compared to rats in vivo. Penetration and retention of Avidin within cartilage is greatly enhanced by electrostatic interactions. Medial tibial cartilage was the thickest of rabbit cartilages, which generated the longest intra-cartilage half-life of Avidin (τ1/2 = 154 h). In contrast, Avidin half-life in thinner rat cartilage was 5-6 times shorter (τ1/2 ∼ 29 h). While a weak correlation (R(2) = 0.43) was found between Avidin half-lives and rabbit tissue GAG concentrations, this correlation improved dramatically (R(2) = 0.96) when normalized to the square of cartilage thickness, consistent with the importance of cartilage thickness to evaluation of drug delivery and retention.


Assuntos
Avidina/farmacocinética , Cartilagem Articular/metabolismo , Sistemas de Liberação de Medicamentos , Animais , Avidina/administração & dosagem , Cartilagem Articular/anatomia & histologia , Feminino , Glicosaminoglicanos/metabolismo , Injeções Intra-Articulares , Modelos Animais , Coelhos , Xantenos
17.
J Orthop Res ; 32(8): 1044-51, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24753019

RESUMO

Intra-articular (i.a.) drug delivery for local treatment of osteoarthritis remains inadequate due to rapid clearance by the vasculature or lymphatics. Local therapy targeting articular cartilage is further complicated by its dense meshwork of collagen and negatively charged proteoglycans, which can prevent even nano-sized solutes from entering. In a previous in vitro study, we showed that Avidin, due to its size (7 nm diameter) and high positive charge (pI 10.5), penetrated the full thickness of bovine cartilage and was retained for 15 days. With the goal of using Avidin as a nano-carrier for cartilage drug delivery, we investigated its transport properties within rat knee joints. Avidin penetrated the full thickness of articular cartilage within 6 h, with a half-life of 29 h, and stayed inside the joint for 7 days after i.a. injection. The highest concentration of Avidin was found in cartilage, the least in patellar tendon and none in the femoral bone; in contrast, negligible Neutravidin (neutral counterpart of Avidin) was present in cartilage after 24 h. A positive correlation between tissue sGAG content and Avidin uptake (R(2) = 0.83) confirmed the effects of electrostatic interactions. Avidin doses up to at least 1 µM did not affect bovine cartilage explant cell viability, matrix catabolism or biosynthesis.


Assuntos
Avidina/administração & dosagem , Cartilagem Articular/metabolismo , Portadores de Fármacos/administração & dosagem , Articulação do Joelho/metabolismo , Animais , Avidina/farmacocinética , Avidina/farmacologia , Cartilagem Articular/efeitos dos fármacos , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Meia-Vida , Injeções Intra-Articulares , Ligamentos/metabolismo , Ratos , Ratos Endogâmicos F344 , Eletricidade Estática , Tendões/metabolismo
18.
Stem Cell Res Ther ; 5(4): 104, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25163844

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) have the ability to repair and regenerate tissue, home to sites of inflammation, and evade the host immune system. As such, they represent an attractive therapy for the treatment of autoimmune inflammatory diseases. However, results from in vivo murine studies in inflammatory arthritis have been conflicting, and this may be due to the genetic background of the MSCs used. It is known that the inflammatory milieu may influence properties of MSCs and that, in the case of human bone marrow-derived MSCs, this may be mediated by the nuclear factor-kappa-B (NF-κB) pathway. We sought to determine whether pro-inflammatory cytokines altered the differentiation and migration capacity of murine MSCs from different mouse strains and whether this was mediated by NF-κB. METHODS: The differentiation and migration of FVB and BALB/c MSCs were carried out in the presence of varying concentrations of tumor necrosis factor-alpha (TNFα) and interleukin (IL)-1ß, and the NF-κB pathway was inhibited in one of two ways: either by transduction of MSCs with an adenoviral vector expressing a super-repressor of NF-κB or by the addition of curcumin to culture media. RESULTS: Both BALB/c and FVB MSCs were sensitive to the effect of pro-inflammatory cytokines in vitro. TNFα and IL-1ß suppressed BALB/c osteogenesis and adipogenesis and FVB osteogenesis. The migration of both cell types toward media containing fetal bovine serum was augmented by pre-stimulation with either cytokine. In neither cell type were the cytokine effects reversed by abrogation of the NF-κB pathway. CONCLUSIONS: These data show that murine MSCs from different genetic backgrounds may be influenced by an inflammatory milieu in a manner that is not mediated by NF-κB, as is the case for human MSCs. This is not mediated by NF-κB. These findings are important and should influence how in vivo trials of murine MSCs are interpreted and the future development of pre-clinical studies in inflammatory diseases.


Assuntos
Diferenciação Celular/genética , Movimento Celular/genética , Interleucina-1beta/farmacologia , Células-Tronco Mesenquimais/citologia , NF-kappa B/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Técnicas de Cultura de Células , Curcumina/farmacologia , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , NF-kappa B/genética , NF-kappa B/metabolismo , Osteogênese/genética , Transdução Genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Cartilage ; 4(1): 63-74, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24533173

RESUMO

OBJECTIVE: Dexamethasone (Dex) is a synthetic glucocorticoid that has pro-anabolic and anti-catabolic effects in cartilage tissue engineering systems, though the mechanisms by which these effects are mediated are not well understood. We tested the hypothesis that the addition of Dex to chondrogenic medium would affect matrix production and aggrecanase activity of human and bovine bone marrow stromal cells (BMSCs) cultured in self-assembling peptide and agarose hydrogels. DESIGN: We cultured young bovine and adult human BMSCs in (RADA)4 self-assembling peptide and agarose hydrogels in medium containing TGF-ß1±Dex and analyzed extracellular matrix composition, aggrecan cleavage products, and the effects of the glucocorticoid receptor antagonist RU-486 on proteoglycan content, synthesis, and catabolic processing. RESULTS: Dex improved proteoglycan synthesis and retention in agarose hydrogels seeded with young bovine cells, but decreased proteoglycan accumulation in peptide scaffolds. These effects were mediated by the glucocorticoid receptor. Adult human BMSCs showed minimal matrix accumulation in agarose, but accumulated ~50% as much proteoglycan and collagen as young bovine BMSCs in peptide hydrogels. Dex reduced aggrecanase activity in (RADA)4 and agarose hydrogels, as measured by anti-NITEGE Western blotting, for both bovine and human BMSC-seeded gels. CONCLUSIONS: The effects of Dex on matrix production are dependent on cell source and hydrogel identity. This is the first report of Dex reducing aggrecanase activity in a tissue engineering culture system.

20.
J Orthop Res ; 30(7): 1095-102, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22213093

RESUMO

Large, osseous, segmental defects heal poorly. Muscle has a propensity to form bone when exposed to an osteogenic stimulus such as that provided by transfer and expression of cDNA encoding bone morphogenetic protein-2 (BMP-2). The present study evaluated the ability of genetically modified, autologous muscle to heal large cranial defects in rats. Autologous grafts (8 mm × 2 mm) were punched from the biceps femoris muscle and transduced intraoperatively with recombinant adenovirus vector containing human BMP-2 or green fluorescent protein cDNA. While the muscle biopsies were incubating with the vector, a central parietal 8 mm defect was surgically created in the calvarium of the same animal. The gene-activated muscle graft was then implanted into the cranial defect. After 8 weeks, crania were examined radiographically, histologically, and by micro-computed tomography and dual energy X-ray absorptiometry. Although none of the defects were completely healed in this time, muscle grafts expressing BMP-2 deposited more than twice as much new bone as controls. Histology confirmed the anatomical integrity of the newly formed bone, which was comparable in thickness and mineral density to the original cranial bone. This study confirms the in vivo osteogenic properties of genetically modified muscle and suggests novel strategies for healing bone.


Assuntos
Doenças Ósseas/terapia , Proteína Morfogenética Óssea 2/genética , Regeneração Óssea/fisiologia , Terapia Genética/métodos , Músculo Esquelético/transplante , Crânio/fisiologia , Adenoviridae/genética , Animais , Diferenciação Celular/fisiologia , Masculino , Músculo Esquelético/citologia , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteogênese/fisiologia , Ratos , Ratos Endogâmicos F344 , Transgenes/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA