Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Eur Cell Mater ; 44: 43-55, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35976149

RESUMO

The interphase between tendon and bone consists of a highly specialised tissue called enthesis. Typically, the enthesis is described as a succession of four different zones: tendon, non-mineralised fibrocartilage, mineralised fibrocartilage and bone. However, the microstructure of the entheses, cellular composition and mechanical properties vary depending on their anatomical location. The present study aimed to characterise three of the most relevant sites of enthesis injury in a rat model: the patellar tendon, the Achilles tendon and the supraspinatus enthesis, in terms of biomechanics, histology and genetic expression. The patellar enthesis presented the highest ultimate load and lowest stiffness of the three, while the supraspinatus was the weakest and stiffest. The histological characterisation revealed key differences at the insertion site for each enthesis. The patellar enthesis showed a large cartilaginous area at the tendon-to-bone interphase whilst this interphase was smaller in the supraspinatus entheses samples. Furthermore, the Achilles tendon enthesis displayed a more abrupt transition from tendon to bone. Additionally, each enthesis exhibited a particular and distinct pattern of expression of tenogenic, chondrogenic and osteogenic markers. This study provided valuable insights for a better understanding of the three entheses at relevant anatomical sites. Moreover, the larger cross-sectional area of the patellar enthesis, the strong mechanical properties and the easier surgical access to this location led to the conclusion that the patellar tendon enthesis site could be most suitable for the development of a preclinical model for general enthesis regeneration studies in rats.


Assuntos
Tendão do Calcâneo , Fibrocartilagem , Tendão do Calcâneo/patologia , Animais , Osso e Ossos , Osteogênese , Ratos , Manguito Rotador
2.
Eur Cell Mater ; 43: 153-161, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35446434

RESUMO

The Achilles tendon is the strongest tendon in the human body but its mechanical behaviour during failure has been little studied and the basis of its high tensile strength has not been elucidated in detail. In the present study, healthy, human, Achilles tendons were loaded to failure in an anatomically authentic fashion while the local deformation and strains were studied in real time, with very high precision, using digital image correlation (DIC). The values determined for the strength of the Achilles tendon were at the high end of those reported in the literature, consistent with the absence of a pre-existing tendinopathy in the samples, as determined by careful gross inspection and histology. Early in the loading cycle, the proximal region of the tendon accumulated high lateral strains while longitudinal strains remained low. However, immediately before rupture, the mid-substance of the Achilles tendon, its weakest part, started to show high longitudinal strains. These new insights advance the understanding of the mechanical behaviour of tendons as they are stretched to failure.


Assuntos
Tendão do Calcâneo , Tendinopatia , Fenômenos Biomecânicos , Humanos , Técnicas In Vitro , Ruptura
3.
Eur Cell Mater ; 41: 668-679, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34111297

RESUMO

The present review acknowledges the tremendous impact of Stephan Perren's strain theory, considered with respect to the earlier contributions of Roux and Pauwels. Then, it provides further insight by examining how the concept of reverse dynamisation extended Perren's theory within a modern context. A key factor of this more contemporary theory is that it introduces variable mechanical conditions at different time points during bone healing, opening the possibility of manipulating biology through mechanics to achieve the desired clinical outcome. The discussion focusses on the current state of the art and the most recent advances made towards optimising and accelerating bone regeneration, by actively controlling the mechanical environment as healing progresses. Reverse dynamisation utilises a very specific mechanical manipulation regimen, with conditions initially flexible to encourage and expedite early callus formation. Once callus has formed, the mechanical conditions are intentionally modified to create a rigid environment under which the soft callus is quickly converted to hard callus, bridging the fracture site and leading to a more rapid union. The relevant literature, principally animal studies, was surveyed to provide ample evidence in support of the effectiveness of reverse dynamisation. By providing a modern perspective on Stephan Perren's strain theory, reverse dynamisation perhaps holds the key to tipping the balance in favour of a more rapid and reliable union when treating acute fractures, osteotomies, non-unions and other circumstances where it is necessary to regenerate bone.


Assuntos
Regeneração Óssea/fisiologia , Osso e Ossos/fisiopatologia , Consolidação da Fratura/fisiologia , Fraturas Ósseas/fisiopatologia , Animais , Calo Ósseo/fisiopatologia , Humanos
4.
Eur Cell Mater ; 41: 345-354, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33729540

RESUMO

Clinical management of delayed healing or non-union of long bone fractures and segmental defects poses a substantial orthopaedic challenge. There are suggestions in the literature that bone healing may be enhanced by inhibiting the activities of T and B lymphocytes, but this remains controversial. To examine this matter in more detail, sub-critical-sized segmental defects were created in the femora of mice and it was assessed whether there might be a benefit from the administration of a Food and Drug Administration (FDA)-approved drug that blocks T cell activation (tacrolimus). Defects were stabilised using an internal plate. In certain groups of animals, 1 mg/kg or 10 mg/kg tacrolimus was delivered locally to the defect site for 3 or 7 d using an implanted osmotic pump with a silicon catheter directing drug delivery into the defect area. Healing was monitored by weekly X-ray and assessed at 12 weeks by mechanical testing, µCT and histology. Radiographic and histological evaluations revealed that 100 % of defects healed well regardless of tacrolimus dosage or duration. A comparison of healed C57BL/6 and Rag1-/- femora by µCT and ex vivo torsion testing showed no differences within mouse strains in terms of bone volume, tissue volume, bone volume/tissue volume ratio, shear modulus, torsional rigidity or torsional stiffness. These data failed to support an important role for tacrolimus in modulating the natural healing of segmental defects under those experimental conditions.


Assuntos
Consolidação da Fratura/efeitos dos fármacos , Fraturas Ósseas/tratamento farmacológico , Fraturas Ósseas/metabolismo , Proteínas de Homeodomínio/metabolismo , Tacrolimo/farmacologia , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Fêmur , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteotomia/métodos , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Microtomografia por Raio-X/métodos
5.
Eur Cell Mater ; 40: 160-171, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33021330

RESUMO

There is much interest in understanding the influence of the immune system on bone healing, including a number of reports suggesting a beneficial effect of FK506 (tacrolimus) in this regard. The influence of FK506 in a rat, femoral, critical size defect was examined using locally implanted, recombinant, human (rh) BMP-2 and adenovirally-transduced, autologous, adipose-derived mesenchymal stromal cells (AD-MSCs) expressing BMP-2. FK506 was delivered systemically using an implanted osmotic pump. Empty defects and those implanted with unmodified AD-MSCs did not heal in the presence or absence of FK506. Defects treated with rhBMP-2 healed with a large callus containing thin cortices and wispy trabeculae; this, too, was unaffected by FK506. A third of defects implanted with adenovirally-transduced AD-MSCs healed, but this improved to 100 % in the presence of FK506. New bone formed in response to BMP-2 synthesised endogenously by the genetically modified cells had a slimmer callus than those healed by rhBMP-2, with improved cortication and advanced reconstitution of marrow. These results suggest that FK506 may have had little effect on the intrinsic biology of bone healing, but improved healing in response to adenovirally-transduced cells by inhibiting immune responses to the first-generation adenovirus used here. Because the genetically modified cells produced bone of higher quality at far lower doses of BMP-2, this approach should be explored in subsequent research.


Assuntos
Diáfises/patologia , Fêmur/patologia , Tacrolimo/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 2/metabolismo , Diáfises/diagnóstico por imagem , Diáfises/efeitos dos fármacos , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fibrina/metabolismo , Masculino , Ratos Endogâmicos F344 , Torção Mecânica
6.
BMC Musculoskelet Disord ; 21(1): 109, 2020 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-32066427

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) based-treatments of cartilage injury are promising but impaired by high levels of hypertrophy after chondrogenic induction with several bone morphogenetic protein superfamily members (BMPs). As an alternative, this study investigates the chondrogenic induction of MSCs via adenoviral gene-delivery of the transcription factor SOX9 alone or in combination with other inducers, and comparatively explores the levels of hypertrophy and end stage differentiation in a pellet culture system in vitro. METHODS: First generation adenoviral vectors encoding SOX9, TGFB1 or IGF1 were used alone or in combination to transduce human bone marrow-derived MSCs at 5 × 102 infectious particles/cell. Thereafter cells were placed in aggregates and maintained for three weeks in chondrogenic medium. Transgene expression was determined at the protein level (ELISA/Western blot), and aggregates were analysed histologically, immunohistochemically, biochemically and by RT-PCR for chondrogenesis and hypertrophy. RESULTS: SOX9 cDNA was superior to that encoding TGFB1, the typical gold standard, as an inducer of chondrogenesis in primary MSCs as evidenced by improved lacuna formation, proteoglycan and collagen type II staining, increased levels of GAG synthesis, and expression of mRNAs associated with chondrogenesis. Moreover, SOX9 modified aggregates showed a markedly lower tendency to progress towards hypertrophy, as judged by expression of the hypertrophy markers alkaline phosphatase, and collagen type X at the mRNA and protein levels. CONCLUSION: Adenoviral SOX9 gene transfer induces chondrogenic differentiation of human primary MSCs in pellet culture more effectively than TGFB1 gene transfer with lower levels of chondrocyte hypertrophy after 3 weeks of in vitro culture. Such technology might enable the formation of more stable hyaline cartilage repair tissues in vivo.


Assuntos
Adenoviridae/genética , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Técnicas de Transferência de Genes , Células-Tronco Mesenquimais/fisiologia , Fatores de Transcrição SOX9/genética , Adulto , Idoso , Células Cultivadas , Feminino , Humanos , Hipertrofia , Masculino , Pessoa de Meia-Idade , Fatores de Transcrição SOX9/biossíntese
7.
Eur Cell Mater ; 38: 14-22, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31332771

RESUMO

Bone autografting remains the clinical model of choice for resolving problematic fractures. The precise mechanisms through which the autograft promotes bone healing are unknown. The present study examined the hypothesis that cells within the autograft secrete osteogenic factors promoting the differentiation of mesenchymal stem cells (MSCs) into osteoblasts. Particles of human bone ("chips") were recovered at the time of joint replacement surgery and placed in culture. Then, conditioned media were added to cultures of human, adipose-derived MSCs under both basal and osteogenic conditions. Contrary to expectation, medium conditioned by bone chips reduced the expression of alkaline phosphatase and strongly inhibited mineral deposition by MSCs cultured in osteogenic medium. Real time PCR revealed the inhibition of collagen type I alpha 1 chain (Col1A1) and osteopontin (OPN) expression. These data indicated that the factors secreted by bone chips inhibited the osteogenic differentiation of MSCs. However, in late cultures, bone morphogenetic protein-2 (BMP-2) expression was stimulated, suggesting the possibility of a delayed, secondary osteogenic effect.


Assuntos
Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteogênese , Comunicação Parácrina , Tecido Adiposo/citologia , Adulto , Idoso , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Osteoblastos/metabolismo , Osteopontina/genética , Osteopontina/metabolismo
9.
J Biomech ; 81: 52-57, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30293824

RESUMO

Rodent models are commonly used to investigate tendon healing, with the biomechanical and structural properties of the healed tendons being important outcome measures. Tendon storage for later testing becomes necessary when performing large experiments with multiple time-points. However, it is unclear whether freezing rodent tendons affects their material properties. Thus the aim of this study was to determine whether freezing rat Achilles tendons affects their biomechanical or structural properties. Tendons were frozen at either -20 °C or -80 °C directly after harvesting, or tested when freshly harvested. Groups of tendons were subjected to several freeze-thaw cycles (1, 2, and 5) within 3 months, or frozen for 9 months, after which the tendons were subjected to biomechanical testing. Additionally, fresh and thawed tendons were compared morphologically, histologically and by transmission electron microscopy. No major differences in biomechanical properties were found between fresh tendons and those frozen once or twice at -20 °C or -80 °C. However, deterioration of tendon properties was found for 5-cycle groups and both long-term freezing groups; after 9 months of freezing at -80 °C the tear resistance of the tendon was reduced from 125.4 ±â€¯16.4N to 74.3 ±â€¯18.4N (p = 0.0132). Moreover, tendons stored under these conditions showed major disruption of collagen fibrils when examined by transmission electron microscopy. When examined histologically, fresh samples exhibited the best cellularity and proteoglycan content of the enthesis. These properties were preserved better after freezing at -80 °C than after freezing at -20 °C, which resulted in markedly smaller chondrocytes and less proteoglycan content. Overall, the best preservation of histological integrity was seen with tendons frozen once at -80 °C. In conclusion, rat Achilles tendons can be frozen once or twice for short periods of time (up to 3 months) at -20 °C or -80 °C for later testing. However, freezing for 9 months at either -20 °C or -80 °C leads to deterioration of certain parameters.


Assuntos
Tendão do Calcâneo/fisiologia , Congelamento , Tendão do Calcâneo/ultraestrutura , Animais , Fenômenos Biomecânicos , Masculino , Microscopia Eletrônica de Transmissão , Ratos Sprague-Dawley , Preservação de Tecido
10.
Eur Cell Mater ; 35: 318-334, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29897097

RESUMO

Several growth factors (GFs) are expressed as tendons heal, but it remains unknown whether their combined application enhances the healing process. This matter was addressed by applying a combination of basic fibroblast growth factor (bFGF), bone morphogenetic protein 12 (BMP-12) and transforming growth factor beta 1 (TGFß1) in a rat Achilles tendon transection model. GFs were applied in one of the three following ways: i) direct application of all three factors at the time of surgery; ii) sequential, tiered percutaneous injection of individual factors immediately after surgery, 48 h and 96 h later; iii) load of all three factors onto a collagen sponge implanted at the time of surgery. After 1, 2, 4 and 8 weeks, healing was assessed based on tendon length and thickness, mechanical strength, stiffness and histology. Best results were achieved when GFs were loaded onto a collagen sponge - with a rapid increase in mechanical strength (load to failure, 71.2 N vs. 7.7 N in controls), consistent tendon length over time (9.9 mm vs. 16.2 mm in controls) and faster tendon remodelling, as measured by histology - followed by tiered injection therapy over 96 h. In conclusion, implantation of a GF-loaded collagen sponge could provide a promising treatment, especially in high-performance athletes and revision cases prone to re-rupture. For conservative treatment, tiered percutaneous GF application could be an option for improving clinical outcome.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Tendões/patologia , Fator de Crescimento Transformador beta1/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Fenômenos Biomecânicos , Colágeno/metabolismo , Cavalos , Masculino , Ratos Sprague-Dawley , Tendões/cirurgia , Suporte de Carga
11.
Am J Surg ; 213(2): 268-272, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27839688

RESUMO

INTRODUCTION: Today's medical learners are Millennials, and reportedly, multitasking pros. We aim to evaluate effect of multitasking on cognitive and technical skills. MATERIALS AND METHODS: 16 medical students completed a mock page and laceration closure separately on day 1 and day 13, and in parallel on day 14. Suturing was graded using GRS and mock pages scored. Total time, suturing and loading times, and percent correct on mock page were compared. RESULTS: Percent correct on mock page improved from days 1-13 and 14 (p < 0.01 and 0.04). GRS improved from days 1-13 and 14 (p = 0.04 and <0.01). Total time suturing was similar on all days. However, time suturing during the mock page on day 14 was prolonged compared to before mock page (p = 0.01). CONCLUSIONS: Medical students can complete cognitive and technical tasks in parallel, without compromising acceptability. However, multitasking results in longer times to complete the complex component of the technical task.


Assuntos
Atenção , Cognição , Lacerações/cirurgia , Estudantes de Medicina , Análise e Desempenho de Tarefas , Humanos , Treinamento por Simulação , Suturas , Telefone , Fatores de Tempo
14.
Eur Cell Mater ; 30: 118-30; discussion 130-1, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26388615

RESUMO

Large segmental defects in bone fail to heal and remain a clinical problem. Muscle is highly osteogenic, and preliminary data suggest that autologous muscle tissue expressing bone morphogenetic protein-2 (BMP-2) efficiently heals critical size defects in rats. Translation into possible human clinical trials requires, inter alia, demonstration of efficacy in a large animal, such as the sheep. Scale-up is fraught with numerous biological, anatomical, mechanical and structural variables, which cannot be addressed systematically because of cost and other practical issues. For this reason, we developed a translational model enabling us to isolate the biological question of whether sheep muscle, transduced with adenovirus expressing BMP-2, could heal critical size defects in vivo. Initial experiments in athymic rats noted strong healing in only about one-third of animals because of unexpected immune responses to sheep antigens. For this reason, subsequent experiments were performed with Fischer rats under transient immunosuppression. Such experiments confirmed remarkably rapid and reliable healing of the defects in all rats, with bridging by 2 weeks and remodelling as early as 3-4 weeks, despite BMP-2 production only in nanogram quantities and persisting for only 1-3 weeks. By 8 weeks the healed defects contained well-organised new bone with advanced neo-cortication and abundant marrow. Bone mineral content and mechanical strength were close to normal values. These data demonstrate the utility of this model when adapting this technology for bone healing in sheep, as a prelude to human clinical trials.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea/genética , Osso e Ossos/lesões , Osso e Ossos/metabolismo , Consolidação da Fratura/genética , Músculo Esquelético/metabolismo , Animais , Animais Geneticamente Modificados , Proteína Morfogenética Óssea 2/genética , Terapia Genética , Vetores Genéticos/uso terapêutico , Masculino , Ratos , Ovinos , Fator de Crescimento Transformador beta/genética
15.
Eur Cell Mater ; 23: 289-98; discussion 299, 2012 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-22522283

RESUMO

Very little is known about the influence of the mechanical environment on the healing of large segmental defects. This partly reflects the lack of standardised, well characterised technologies to enable such studies. Here we report the design, construction and characterisation of a novel external fixator for use in conjunction with rat femoral defects. This device not only imposes a predetermined axial stiffness on the lesion, but also enables the stiffness to be changed during the healing process. The main frame of the fixator consists of polyethylethylketone with titanium alloy mounting pins. The stiffness of the fixator is determined by interchangeable connection elements of different thicknesses. Fixators were shown to stabilise 5 mm femoral defects in rats in vivo for at least 8 weeks during unrestricted cage activity. No distortion or infections, including pin infections, were noted. The healing process was simulated in vitro by inserting into a 5 mm femoral defect, materials whose Young's moduli approximated those of the different tissues present in regenerating bone. These studies confirmed that, although the external fixator is the major determinant of axial stiffness during the early phase of healing, the regenerate within the lesion subsequently dominates this property. There is much clinical interest in altering the mechanics of the defect to enhance bone healing. Our data suggest that, if alteration of the mechanical environment is to be used to modulate the healing of large segmental defects, this needs to be performed before the tissue properties become dominant.


Assuntos
Fixadores Externos/normas , Fraturas do Fêmur/cirurgia , Fêmur/cirurgia , Fixação de Fratura/instrumentação , Ligas/química , Ligas/normas , Animais , Pinos Ortopédicos/normas , Força Compressiva , Elasticidade , Desenho de Equipamento , Fraturas do Fêmur/fisiopatologia , Fêmur/lesões , Fêmur/fisiopatologia , Masculino , Teste de Materiais , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Titânio/química , Cicatrização/fisiologia
16.
Adv Drug Deliv Rev ; 64(12): 1331-40, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22480730

RESUMO

Gene delivery to bone is useful both as an experimental tool and as a potential therapeutic strategy. Among its advantages over protein delivery are the potential for directed, sustained and regulated expression of authentically processed, nascent proteins. Although no clinical trials have been initiated, there is a substantial pre-clinical literature documenting the successful transfer of genes to bone, and their intraosseous expression. Recombinant vectors derived from adenovirus, retrovirus and lentivirus, as well as non-viral vectors, have been used for this purpose. Both ex vivo and in vivo strategies, including gene-activated matrices, have been explored. Ex vivo delivery has often employed mesenchymal stem cells (MSCs), partly because of their ability to differentiate into osteoblasts. MSCs also have the potential to home to bone after systemic administration, which could serve as a useful way to deliver transgenes in a disseminated fashion for the treatment of diseases affecting the whole skeleton, such as osteoporosis or osteogenesis imperfecta. Local delivery of osteogenic transgenes, particularly those encoding bone morphogenetic proteins, has shown great promise in a number of applications where it is necessary to regenerate bone. These include healing large segmental defects in long bones and the cranium, as well as spinal fusion and treating avascular necrosis.


Assuntos
Regeneração Óssea/genética , Osso e Ossos/metabolismo , Terapia Genética/métodos , Animais , Doenças Ósseas/patologia , Doenças Ósseas/terapia , Proteínas Morfogenéticas Ósseas/administração & dosagem , Osso e Ossos/patologia , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Transplante de Células-Tronco Mesenquimais/métodos
17.
J Cell Physiol ; 227(2): 416-20, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21948071

RESUMO

Orthopedic gene therapy has been the topic of considerable research for two decades. The preclinical data are impressive and many orthopedic conditions are well suited to genetic therapies. But there have been few clinical trials and no FDA-approved product exists. This paper examines why this is so. The reasons are multifactorial. Clinical translation is expensive and difficult to fund by traditional academic routes. Because gene therapy is viewed as unsafe and risky, it does not attract major funding from the pharmaceutical industry. Start-up companies are burdened by the complex intellectual property environment and difficulties in dealing with the technology transfer offices of major universities. Successful translation requires close interactions between scientists, clinicians and experts in regulatory and compliance issues. It is difficult to create such a favorable translational environment. Other promising fields of biological therapy have contemplated similar frustrations approximately 20 years after their founding, so there seem to be more general constraints on translation that are difficult to define. Gene therapy has noted some major clinical successes in recent years, and a sense of optimism is returning to the field. We hope that orthopedic applications will benefit collaterally from this upswing and move expeditiously into advanced clinical trials.


Assuntos
Terapia Genética/métodos , Doenças Musculoesqueléticas/genética , Doenças Musculoesqueléticas/terapia , Ortopedia/métodos , Ensaios Clínicos como Assunto , Humanos , Doenças Musculoesqueléticas/patologia , Medicina Regenerativa , Engenharia Tecidual
18.
Gene Ther ; 16(8): 944-52, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19675584

RESUMO

Gene therapies directed toward the treatment of arthritis and tissue repair continue to be the most active areas of research for bone and joint diseases. In the past 2 years, two trials in rheumatoid arthritis have been completed. a Phase I study reporting safety and a Phase I/II study that has yet to be published. An additional, small study has reported the first evidence of clinical efficacy. Two Phase I trials of gene therapy for osteoarthritis have also been initiated. There is much preclinical activity in developing AAV vectors for future trials in the gene therapy of arthritis. Research into tissue repair and regeneration remains at the preclinical stage, but a considerable volume of research attests to the promise of gene transfer in this arena, especially in the context of bone healing. For tissue repair, the major research questions are still which genes to use and how best to deliver them.


Assuntos
Doenças Ósseas/terapia , Artropatias/terapia , Artrite/terapia , Ensaios Clínicos como Assunto , Técnicas de Transferência de Genes , Terapia Genética
19.
Arthritis Rheum ; 60(3): 801-12, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19248089

RESUMO

OBJECTIVE: The differentiation of mesenchymal stem cells (MSCs) into chondrocytes provides an attractive basis for the repair and regeneration of articular cartilage. Under clinical conditions, chondrogenesis will often need to occur in the presence of mediators of inflammation produced in response to injury or disease. The purpose of this study was to examine the effects of 2 important inflammatory cytokines, interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha), on the chondrogenic behavior of human MSCs. METHODS: Aggregate cultures of MSCs recovered from the femoral intermedullary canal were used. Chondrogenesis was assessed by the expression of relevant transcripts by quantitative reverse transcription-polymerase chain reaction analysis and examination of aggregates by histologic and immunohistochemical analyses. The possible involvement of NF-kappaB in mediating the effects of IL-1beta was examined by delivering a luciferase reporter construct and a dominant-negative inhibitor of NF-kappaB (suppressor-repressor form of IkappaB [srIkappaB]) with adenovirus vectors. RESULTS: Both IL-1beta and TNFalpha inhibited chondrogenesis in a dose-dependent manner. This was associated with a marked activation of NF-kappaB. Delivery of srIkappaB abrogated the activation of NF-kappaB and rescued the chondrogenic response. Although expression of type X collagen followed this pattern, other markers of hypertrophic differentiation responded differently. Matrix metalloproteinase 13 was induced by IL-1beta in a NF-kappaB-dependent manner. Alkaline phosphatase activity, in contrast, was inhibited by IL-1beta regardless of srIkappaB delivery. CONCLUSION: Cell-based repair of lesions in articular cartilage will be compromised in inflamed joints. Strategies for enabling repair under these conditions include the use of specific antagonists of individual pyrogens, such as IL-1beta and TNFalpha, or the targeting of important intracellular mediators, such as NF-kappaB.


Assuntos
Condrogênese/fisiologia , Interleucina-1beta/fisiologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Idoso , Diferenciação Celular , Células Cultivadas , Colágeno Tipo X/metabolismo , Feminino , Humanos , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/fisiologia
20.
Eur Cell Mater ; 18: 96-111, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-20073015

RESUMO

We report a novel technology for the rapid healing of large osseous and chondral defects, based upon the genetic modification of autologous skeletal muscle and fat grafts. These tissues were selected because they not only possess mesenchymal progenitor cells and scaffolding properties, but also can be biopsied, genetically modified and returned to the patient in a single operative session. First generation adenovirus vector carrying cDNA encoding human bone morphogenetic protein-2 (Ad.BMP-2) was used for gene transfer to biopsies of muscle and fat. To assess bone healing, the genetically modified ("gene activated") tissues were implanted into 5mm-long critical size, mid-diaphyseal, stabilized defects in the femora of Fischer rats. Unlike control defects, those receiving gene-activated muscle underwent rapid healing, with evidence of radiologic bridging as early as 10 days after implantation and restoration of full mechanical strength by 8 weeks. Histologic analysis suggests that the grafts rapidly differentiated into cartilage, followed by efficient endochondral ossification. Fluorescence in situ hybridization detection of Y-chromosomes following the transfer of male donor muscle into female rats demonstrated that at least some of the osteoblasts of the healed bone were derived from donor muscle. Gene activated fat also healed critical sized defects, but less quickly than muscle and with more variability. Anti-adenovirus antibodies were not detected. Pilot studies in a rabbit osteochondral defect model demonstrated the promise of this technology for healing cartilage defects. Further development of these methods should provide ways to heal bone and cartilage more expeditiously, and at lower cost, than is presently possible.


Assuntos
Tecido Adiposo/transplante , Doenças Ósseas/terapia , Doenças das Cartilagens/terapia , Técnicas de Transferência de Genes , Músculo Esquelético/transplante , Transplante de Tecidos/métodos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Regeneração Óssea/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem da Célula/fisiologia , Modelos Animais de Doenças , Feminino , Fêmur/citologia , Fêmur/metabolismo , Fêmur/cirurgia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Terapia Genética/métodos , Vetores Genéticos/genética , Sobrevivência de Enxerto/fisiologia , Humanos , Masculino , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Coelhos , Ratos , Ratos Endogâmicos F344 , Transplante Autólogo/métodos , Resultado do Tratamento , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA