Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Ther Methods Clin Dev ; 29: 350-363, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37214314

RESUMO

Bone morphogenetic protein-2 (BMP-2) is an osteogenic protein used clinically to enhance bone healing. However, it must be applied in very high doses, causing adverse side effects and increasing costs while providing only incremental benefit. Preclinical models of bone healing using gene transfer to deliver BMP-2 suggest that transgenic BMP-2 is much more osteogenic than rhBMP-2. Using a reporter mesenchymal cell line, we found transgenic human BMP-2 cDNA to be at least 100-fold more effective than rhBMP-2 in signaling. Moreover, a substantial portion of the BMP-2 produced by the transduced cells remained cell associated. Signaling by transgenic BMP-2 occurred via binding to the type I receptor, activating the associated kinase and generating phospho-smads. Signaling was partially resistant to noggin, an important extracellular inhibitor of BMP-2, possibly because nascent BMP-2 binds to its cell surface receptor during secretion and thus signals in a protected peri-cellular environment. Although the amounts of BMP-2 secreted by the transduced cells were too low to affect distant cells, transduced cells were able to induce signaling in a paracrine fashion that required close proximity of the cells, possibly cell-to-cell contact. The greater osteogenic potency of transgenic BMP-2 was confirmed with human bone marrow stromal cells.

2.
J Orthop Res ; 41(9): 1934-1944, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36850029

RESUMO

This study defined and compared the course of native, impaired and growth factor-stimulated bone regeneration in a rat femoral defect model. A mid-diaphyseal defect with rigid internal fixation was surgically created in the right femur of male Fischer rats and serially analyzed over 36 weeks. Native bone regeneration was modeled using a sub-critical, 1 mm size defect, which healed uneventfully. Critical size defects of 5 mm were used to analyze impaired bone regeneration. In a third group, the 5 mm defects were filled with 11 µg of recombinant human bone morphogenetic protein 2 (rhBMP2) impregnated onto an absorbable collagen sponge, modeling its clinical use. Native bone regeneration was characterized by endochondral ossification with progressive remodeling to ultimately resemble intact femora. An endochondral response was also observed under conditions of impaired bone regeneration, but by week 8 medullary capping occurred with fibrofatty consolidation of the tissue within the defect, resembling an atrophic non-union. rhBMP2 treatment was associated with prolonged inflammatory cytokine expression and rapid intramembranous bone formation occurring with reduced expression of cartilage-associated collagens. Between weeks 4 and 36, rhBMP2-treated bones demonstrated decreased trabecular number and increased trabecular separation, which resulted in inferior mechanical properties compared with bones that healed naturally. Clinical Significance: Recombinant human bone morphogenetic protein 2 (rhBMP2) is used clinically to promote healing of long bones. Our data suggest that it drives intramembraneous ossification producing an inferior regenerate that deteriorates with time. Clinical outcomes would be improved by technologies favoring endochondral regenerative ossification.


Assuntos
Proteína Morfogenética Óssea 2 , Regeneração Óssea , Ratos , Humanos , Masculino , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/uso terapêutico , Cicatrização , Fêmur , Osteogênese , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
3.
Gene Ther ; 30(7-8): 587-591, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260797

RESUMO

Osteoarthritis (OA) is a leading cause of disability with no cure and only supportive therapy. Adeno-associated virus (AAV) serotype 2.5 is being used in a Phase I clinical trial to deliver the interleukin-1 receptor antagonist into knee joints with OA. Neutralizing antibodies (Nab) directed against AAV2.5, if present, could inhibit gene transfer. Here, we report the prevalence of AAV2.5 Nab in the sera and synovial fluids of patients with OA. Nab titers were measured by their ability to inhibit in vitro transduction by AAV2.5 encoding GFP. Of 44 synovial fluids from patients with mid-stage and advanced OA, 43% had undetectable Nab; 25% had low titers (<1:100), 16% had medium titers (1:100-1:1000) and 16% had high titers (>1:1000) of Nab. Titers of AAV2.5 Nabs correlated with those of AAV2, but not with those of AAV5. Serum titers of AAV2.5 Nab correlated positively with titers in synovial fluid, and were never less than the matched synovial fluid titers. These findings suggest that high titers of Nab against AAV2.5 are uncommon in the synovial fluids of patients with OA, and individuals with high synovial fluid Nab titers can be identified by measuring titers in the serum.


Assuntos
Anticorpos Neutralizantes , Osteoartrite , Humanos , Líquido Sinovial , Anticorpos Antivirais , Prevalência , Vetores Genéticos/genética , Terapia Genética , Osteoartrite/terapia , Dependovirus/genética
4.
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
5.
Osteoarthr Cartil Open ; 4(2)2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36338933

RESUMO

Objective: To evaluate a single-step, gene-based procedure for repairing osteochondral lesions. Design: Osteochondral lesions were created in the patellar groove of skeletally mature rabbits. Autologous bone marrow aspirates were mixed with adenovirus vectors carrying cDNA encoding green fluorescent protein (Ad.GFP) or transforming growth factor-ß1 (Ad.TGF-ß1) and allowed to clot. The clotted marrow was press-fit into the defects. Animals receiving Ad.GFP were euthanized at 2 weeks and intra-articular expression of GFP examined by fluorescence microscopy. Animals receiving Ad.TGF-ß1 were euthanized at 3 months and 12 months; repair was compared to empty defects using histology and immunohistochemistry. Complementary in vitro experiments assessed transgene expression and chondrogenesis in marrow clots and fibrin gels. In a subsequent pilot study, repair at 3 months using a fibrin gel to encapsulate Ad.TGF-ß1 was evaluated. Results: At 2 weeks, GFP expression was seen at variable levels within the cartilaginous lesion. At 3 months, there was no statistically significant improvement (p > 0.05) in healing of lesions receiving Ad.TGF-ß1 and variability was high. At 12 months, there were still no significant difference (p > 0.05) between the empty defects and those receiving Ad.TGF-ß1 in the overall, cartilage, and bone scores. Variability was still high. In vitro experiments suggested that variability reflected variable transduction efficiency and chondrogenic activity of the marrow clots; using fibrin gels instead of marrow may address this issue but more research is needed. Conclusions: This approach to improving the repair of osteochondral lesions needs further refinement to reduce variability and provide a more robust outcome.

6.
Cells ; 11(11)2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35681447

RESUMO

The use of multiphasic scaffolds to treat injured tendon-to-bone entheses has shown promising results in vitro. Here, we used two versions of a biphasic silk fibroin scaffold to treat an enthesis defect created in a rat patellar model in vivo. One version presented a mixed transition between the bony and the tendon end of the construct (S-MT) while this transition was abrupt in the second version (S-AT). At 12 weeks after surgery, the S-MT scaffold promoted better healing of the injured enthesis, with minimal undesired ossification of the insertion area. The expression of tenogenic and chondrogenic markers was sustained for longer in the S-MT-treated group and the tangent modulus of the S-MT-treated samples was similar to the native tissue at 12 weeks while that of the S-AT-treated enthesis was lower. Our study highlights the important role of the transition zone of multiphasic scaffolds in the treatment of complex interphase tissues such as the tendon-to-bone enthesis.


Assuntos
Fibroínas , Traumatismos dos Tendões , Alicerces Teciduais , Cicatrização , Animais , Fibroínas/farmacologia , Interfase , Ratos , Tendões
7.
Sci Adv ; 8(7): eabl6242, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35171668

RESUMO

Large segmental osseous defects heal poorly. Recombinant, human bone morphogenetic protein-2 (rhBMP-2) is used clinically to promote bone healing, but it is applied at very high doses that cause adverse side effects and raise costs while providing only incremental benefit. We describe a previously unexplored, alternative approach to bone regeneration using chemically modified messenger RNA (cmRNA). An optimized cmRNA encoding BMP-2 was delivered to critical-sized femoral osteotomies in rats. The cmRNA remained orthotopically localized and generated BMP locally for several days. Defects healed at doses ≥25 µg of BMP-2 cmRNA. By 4 weeks, all animals treated with 50 µg of BMP-2 cmRNA had bridged bone defects without forming the massive callus seen with rhBMP-2. Moreover, such defects recovered normal mechanical strength quicker and initiated bone remodeling faster. cmRNA regenerated bone via endochondral ossification, whereas rhBMP-2 drove intramembranous osteogenesis; cmRNA provides an innovative, safe, and highly translatable technology for bone healing.


Assuntos
Proteína Morfogenética Óssea 2 , Regeneração Óssea , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/farmacologia , Fêmur , Osteogênese , RNA Mensageiro/genética , Ratos , Proteínas Recombinantes/farmacologia , Cicatrização
8.
J Histochem Cytochem ; 69(10): 633-643, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34549650

RESUMO

Collagen has a major role in the structural organization of tendons. Picrosirius red (PSR) staining viewed under polarized light microscopy is the standard method to evaluate the organization of collagen fibers in tissues. It is also used to distinguish between type I and type III collagen in tissue sections. However, accurate analysis and interpretation of PSR images are challenging because of technical factors and historical misconceptions. The aim of this study was to clarify whether collagen types I and III can be distinguished by PSR staining in rat Achilles tendons, using double immunohistochemistry as the positive control. Our findings showed that PSR staining viewed with polarized light microscopy was suitable for qualitative and quantitative assessment of total collagen but was not able to distinguish collagen types. We found it critical to use a polarizing microscope equipped with a rotating stage; tendon section orientation at 45° with respect to crossed polarizers was optimal for the qualitative and quantitative assessment of collagen organization. Immunohistochemistry was superior to PSR staining for detection of collagen type III. We also compared formalin and Bouin solution as fixatives. Both produced similar birefringence, but formalin-fixed tendons provided higher quality histological detail with both hematoxylin-eosin and immunostaining.


Assuntos
Compostos Azo/química , Colágeno Tipo III/análise , Colágeno Tipo I/análise , Coloração e Rotulagem , Tendões/química , Animais , Ratos , Ratos Sprague-Dawley
9.
Tissue Eng Part A ; 25(1-2): 131-144, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30009674

RESUMO

IMPACT STATEMENT: The use of chemically modified RNA (cmRNA) with increased stability using translation initiator of short untranslated regions (TISU) offers the prospect of finally allowing us to unlock the potent osteogenic properties of BMP-2 in a clinically expedient manner. As noted, delivery of recombinant BMP-2 protein has had modest clinical efficacy, whereas gene delivery is effective but very difficult to translate into human clinical use. This study shows the great potential of cmRNA encoding BMP-2 with TISU in a long-bone critical-sized rat model.


Assuntos
Proteína Morfogenética Óssea 2 , Técnicas de Transferência de Genes , Células-Tronco Mesenquimais/metabolismo , Osteogênese , RNA Mensageiro , Animais , Proteína Morfogenética Óssea 2/biossíntese , Proteína Morfogenética Óssea 2/genética , Células HEK293 , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Ratos , Ratos Endogâmicos F344
10.
Thyroid ; 25(10): 1145-50, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26213859

RESUMO

BACKGROUND: Activation of thyrotropin receptor (TSHR) and/or insulin-like growth factor (IGF-1) receptor (IGF-1R) enhances HA production and adipogenesis in orbital fibroblasts from patients with Graves' ophthalmopathy (GO) and recapitulates the tissue remodeling characteristic of the orbit in GO. A functional relationship between TSHR and IGF-1R has long been postulated, and recently bidirectional crosstalk between the receptors in GO fibroblasts was demonstrated. Because the transcription factor Forkhead box O-1 (FOXO1) was recently shown to be a critical downstream mediator of TSH and IGF-1 effects on thyrocyte proliferation, studies were designed to determine whether FOXO1 might similarly act as a common mediator of M22, a stimulatory TSHR antibody (TSAb), and IGF-1 in GO orbital fibroblasts. METHODS: FOXO1 mRNA and protein were measured in orbital tissue specimens derived from normal individuals and patients with GO. In addition, the control of FOXO1 cellular localization was investigated using quantitative Western blotting of fractionated cell lysates from orbital fibroblasts treated with M22 and/or IGF-1 with or without specific TSHR, IGF-1R, or PI3K/AKT1/2 inhibitors. RESULTS: Significantly lower levels of both FOXO1 mRNA and protein were found in GO orbital tissue specimens compared with normal orbital tissues (M = 39%, p = 0.043; M = 46.4%; p = 0.028, respectively). In addition, treatment of GO orbital cultures with M22, IGF-1, or M22 plus IGF-1 increased cytoplasmic FOXO1 compared with control (1.63-fold, p = 0.008; 1.68-fold, p = 0.001; 1.61-fold, p ≤ 0.001, respectively) and decreased nuclear FOXO1 (M = 28%, p = 0.002; M = 38%, p ≤ 0.001; M = 35%, p = 0.007, respectively). These effects were inhibited by co-treatment with the respective, but not the opposite, receptor antagonist. AKT inhibition of M22 or IGF-1-treated cultures was found to increase nuclear (1.4-fold, p = 0.026; 1.3-fold, p = 0.001, respectively) and decrease cytoplasmic (24.2%, p = 0.001; 36%, p = 0.004, respectively) FOXO1 localization. CONCLUSIONS: These data point to FOXO1 as an important mediator of TSAb and IGF-1 action via their cognate receptors in GO orbital fibroblasts. These findings provide a link between the low FOXO1 protein levels demonstrated in GO orbital tissue and the tissue remodeling characteristic of GO, and suggest novel therapy for GO aimed at increasing nuclear expression of FOXO1 in GO target cells.


Assuntos
Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Oftalmopatia de Graves/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Receptores da Tireotropina/imunologia , Anticorpos/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Proteína Forkhead Box O1 , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Transdução de Sinais/efeitos dos fármacos
11.
J Clin Endocrinol Metab ; 98(5): 2153-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23482611

RESUMO

CONTEXT: Graves ophthalmopathy (GO) is an autoimmune disorder characterized by increased adipogenesis and hyaluronan (HA) production by orbital fibroblasts. Circulating autoantibodies (thyroid-stimulating antibodies [TSAbs]) directed at the thyrotropin receptor (TSHR) on these cells stimulate or augment these cellular processes. A recently developed drug-like small molecule inverse agonist of TSHR, NCGC00229600, termed 1, binds to TSHR and blocks basal and stimulated signal transduction. OBJECTIVE: The purpose of this article was to determine whether 1 might inhibit HA production and relevant signaling pathways in orbital fibroblasts cultured in the presence of monoclonal TSAbs or bovine TSH (bTSH). DESIGN: Primary cultures of undifferentiated GO orbital fibroblasts (n = 13) were untreated or treated with a TSAb (M22 or MS-1) or bTSH in serum-free medium, with or without 1 or a TSHR neutral antagonist, NCGC00242595, termed 2, which does not inhibit basal signaling but does inhibit stimulated signaling. MAIN OUTCOME MEASURES: cAMP production, Akt phosphorylation (Ser473pAkt in media and immunoblotting for pAkt/total Akt), and HA production were analyzed. RESULTS: Compound 1 inhibited basal cAMP, pAkt, and HA production and that stimulated by M22 in undifferentiated orbital fibroblasts. Inhibition of HA production was dose-dependent, with a half-maximal inhibitory dose of 830 nM. This compound also inhibited MS-1- and bTSH-stimulated cAMP, pAkt, and HA production. Compound 2 did not inhibit basal HA production but did inhibit M22-stimulated HA production. CONCLUSIONS: Because cAMP, pAkt, and HA production are fibroblast functions that are activated via TSHR signaling and are important in the pathogenesis of GO, small molecule TSHR antagonists may prove to be effective in the treatment or prevention of the disease in the future.


Assuntos
Olho/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Oftalmopatia de Graves/tratamento farmacológico , Imunoglobulinas Estimuladoras da Glândula Tireoide/metabolismo , Piridinas/farmacologia , Quinazolinonas/farmacologia , Receptores da Tireotropina/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/imunologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Anticorpos Monoclonais/metabolismo , Desdiferenciação Celular , Células Cultivadas , AMP Cíclico/metabolismo , Agonismo Inverso de Drogas , Olho/imunologia , Olho/metabolismo , Olho/patologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Oftalmopatia de Graves/imunologia , Oftalmopatia de Graves/metabolismo , Oftalmopatia de Graves/patologia , Humanos , Ácido Hialurônico/metabolismo , Concentração Osmolar , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/metabolismo , Tireotropina/agonistas , Tireotropina/antagonistas & inibidores , Tireotropina/farmacologia
12.
J Clin Endocrinol Metab ; 97(5): 1681-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22399503

RESUMO

CONTEXT: Graves' ophthalmopathy (GO) is characterized by expanded volume of the orbital fat and extraocular muscle tissues and elevated levels of TSH receptor autoantibodies (TRAb). The expansion of orbital tissues involves accumulation of hyaluronic acid (HA) within the orbit. OBJECTIVE: The objective of the study was to determine whether a monoclonal stimulatory TRAb (M22) impacts HA synthesis in GO orbital cells and, if so, whether this might be blocked by an IGF-I receptor (IGF-IR)-blocking antibody (1H7) or inhibitors of various downstream signaling cascades. DESIGN: GO orbital fibroblast cultures (n = 6) were treated with M22, bovine TSH (bTSH), or IGF-I in serum-free medium. Some cultures also received 1H7, LY294002, rapamycin, or protein kinase A inhibitor. MAIN OUTCOME MEASURES: HA production and phosphorylated Akt levels in media or immunoblotting for phosphorylated Akt were measured. RESULTS: M22 or bTSH stimulated HA synthesis (2.1-fold with 100 ng/ml M22 and 1.9-fold with 10 U/liter bTSH; P < 0.05 each). M22-induced HA synthesis was inhibited by LY294002 or rapamycin but not by protein kinase inhibitor. HA synthesis stimulated by M22 or IGF-I was inhibited by 1H7 (mean 36.6 ± 5.6% and mean 45.8 ± 7.6%, respectively; P < 0.05 each). Similarly, M22- or IGF-I-stimulated Akt phosphorylation was inhibited by 1H7 (mean 54 ± 9.6 and 36.1 ± 8.8%, respectively; P = 0.01 each). CONCLUSIONS: The stimulatory TRAb M22 increases HA production in undifferentiated GO orbital fibroblasts via phosphoinositide 3-kinase/phosphorylated AKT/mammalian target of rapamycin activation. Blockade of IGF-IR inhibits both HA synthesis and Akt phosphorylation induced by M22 or IGF-I in these cells, suggesting that TSH receptor and IGF-IR signaling may be closely linked in the GO orbit.


Assuntos
Anticorpos Bloqueadores/farmacologia , Fibroblastos/efeitos dos fármacos , Oftalmopatia de Graves/imunologia , Ácido Hialurônico/biossíntese , Receptor IGF Tipo 1/imunologia , Receptores da Tireotropina/imunologia , Anticorpos Bloqueadores/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Receptor IGF Tipo 1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
13.
J Mol Endocrinol ; 46(3): 155-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21321093

RESUMO

Graves' ophthalmopathy (GO) is characterized by expanded volume of the orbital tissues associated with elevated serum levels of TSH receptor (TSHR) autoantibodies. Because previous studies have demonstrated evidence of adipogenesis within the GO orbit, we sought to determine whether M22, a human monoclonal antibody directed against TSHR, enhances adipogenesis in orbital fibroblasts from patients with GO and, if so, to identify signaling mechanisms involved. GO orbital fibroblast cultures (n=10) were treated for 10 days with bovine TSH (1 or 10.0 U/l) or M22 (1 or 10 ng/ml) in serum-free adipocyte differentiation medium. Some cultures also received a phosphoinositide 3-kinase (PI3K) inhibitor or an inhibitor of cAMP production. In other experiments, confluent cultures (n=8) were treated for between 1 and 30  min with TSH (0.1-10.0 U/l) or M22 (0.1-100 ng/ml) with measurement of cAMP production or levels of phosphorylated AKT (pAKT). We found levels of adiponectin, leptin, and TSHR mRNA to be increased in GO cultures treated for 10 days with either M22 (2.6 mean fold ± 0.7; P=0.03) or TSH (13.2 ± 5.8-fold, P=0.048). In other studies, M22 and TSH stimulated cAMP production and pAKT levels in GO cells. Inhibition of PI3K activity during 10 days in culture decreased the levels of M22-stimulated mRNA encoding adiponectin (67 ± 12%; P=0.021), as well as adiponectin and CCAAT/enhancer-binding protein α protein levels. In conclusion, M22 is a pro-adipogenic factor in GO orbital preadipocytes. This antibody appears to act via the PI3K signaling cascade, suggesting that inhibition of PI3K signaling may represent a potential novel therapeutic approach in GO.


Assuntos
Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Oftalmopatia de Graves/metabolismo , Órbita/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores da Tireotropina/agonistas , Adipócitos/metabolismo , Western Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tireotropina/farmacologia
14.
Thyroid ; 20(1): 59-65, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20017620

RESUMO

BACKGROUND: Patients with Graves' ophthalmopathy (GO) have circulating autoantibodies directed against the thyrotropin receptor (TSHR) and elevated levels of the proinflammatory cytokine interleukin-6 (IL-6) in both serum and orbital tissues. We hypothesized that these autoantibodies might increase IL-6 expression and secretion in preadipocyte fibroblasts and adipocytes from patients with GO, and thus directly impact the clinical activity of the disease. METHODS: IL-6 mRNA levels were measured in cultures of GO orbital preadipocytes (n = 3) treated during adipocyte differentiation with a monoclonal stimulatory TSHR antibody (M22; 10 ng/mL), IL-6 (1 ng/mL), or TSH (10 U/L). Additionally, levels of IL-6 protein secretion were assessed after adipocyte differentiation in orbital cultures exposed to TSH or M22 for 24 or 48 hours (n = 8). IL-6 mRNA levels were also measured in orbital adipose tissue specimens from well-characterized GO patients (n = 9) and normal individuals (n = 9). RESULTS: Treatment of GO orbital preadipocyte cultures with IL-6, TSH, or M22 during adipocyte differentiation resulted in increased IL-6 mRNA levels (3.1-fold, 2.9-fold, and 2.7-fold, respectively; p < 0.05). Treatment of orbital cultures with M22 or TSH after adipocyte differentiation enhanced the release of IL-6 protein into the medium at both 24 and 48 hours for TSH (mean 1.9- and 2.3-fold; p = 0.002 and 0.015, respectively) and at 48 hours for M22 (mean 2.0-fold; p = 0.005). In addition, we found mean IL-6 mRNA levels to be significantly increased in GO orbital adipose tissue specimens (10-fold; p < 0.01), primarily attributable to high levels in three of the four patients with clinical activity scores >or=5. CONCLUSIONS: Both TSH and M22 increase IL-6 expression in orbital preadipocyte fibroblasts and IL-6 secretion by mature adipocytes. These results suggest that circulating TSHR autoantibodies in GO might play a direct role in the clinical activity of the disease.


Assuntos
Adipócitos/metabolismo , Autoanticorpos/imunologia , Oftalmopatia de Graves/metabolismo , Interleucina-6/metabolismo , Órbita , Receptores da Tireotropina/imunologia , Tireotropina/farmacologia , Adipócitos/efeitos dos fármacos , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Adulto , Idoso , Anticorpos Monoclonais/imunologia , Diferenciação Celular , Células Cultivadas , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Oftalmopatia de Graves/imunologia , Humanos , Interleucina-6/genética , Interleucina-6/farmacologia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Tireotropina/fisiologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
15.
J Immunol ; 183(6): 4013-20, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19717523

RESUMO

Eosinophil granule proteins are deposited in cutaneous lesions in many human diseases, but how these proteins contribute to pathophysiology is obscure. We injected eosinophil cationic protein (ECP or RNase 3), eosinophil-derived neurotoxin (EDN or RNase 2), eosinophil peroxidase (EPO), and major basic protein-1 (MBP1) intradermally into guinea pig and rabbit skin. ECP and EDN each induced distinct skin lesions at >or=2.5 microM that began at 2 days, peaking at approximately 7 days and persisting up to 6 wk. These lesions were ulcerated (ECP) or crusted (EDN) with marked cellular infiltration. EPO and MBP1 (10 microM) each produced perceptible induration and erythema with moderate cellular infiltration resolving within 2 wk. ECP and EDN localized to dermal cells within 2 days, whereas EPO and MBP1 remained extracellular. Overall, cellular localization and RNase activity of ECP and EDN were critical for lesion formation; differential glycosylation, net cationic charge, or RNase activity alone did not account for lesion formation. Ulcerated lesions from patients with the hypereosinophilic syndrome showed ECP and EDN deposition comparable to that in guinea pig skin. In conclusion, ECP and EDN disrupt skin integrity and cause inflammation. Their presence in ulcerative skin lesions may explain certain findings in human eosinophil-associated diseases.


Assuntos
Proteínas Granulares de Eosinófilos/toxicidade , Eosinófilos/enzimologia , Ribonucleases/toxicidade , Dermatopatias/etiologia , Animais , Proteína Catiônica de Eosinófilo/administração & dosagem , Proteína Catiônica de Eosinófilo/toxicidade , Proteínas Granulares de Eosinófilos/administração & dosagem , Proteína Básica Maior de Eosinófilos/administração & dosagem , Proteína Básica Maior de Eosinófilos/toxicidade , Peroxidase de Eosinófilo/administração & dosagem , Peroxidase de Eosinófilo/toxicidade , Neurotoxina Derivada de Eosinófilo/administração & dosagem , Neurotoxina Derivada de Eosinófilo/toxicidade , Eosinofilia/patologia , Cobaias , Humanos , Coelhos , Ribonucleases/administração & dosagem , Dermatopatias/patologia , Úlcera/etiologia
16.
Thyroid ; 18(12): 1291-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18976167

RESUMO

BACKGROUND: Thy-1 is a surface protein that defines functionally distinct subpopulations of fibroblasts, with those lacking the antigen being capable of adipogenesis. Because increased fat cell development is a hallmark of the orbit in Graves' ophthalmopathy (GO), we wished to compare baseline Thy-1 expression in orbital fibroblasts from GO patients and normal individuals, and determine whether levels of the protein might be impacted by adipogenesis following peroxisome proliferator activator-gamma ligation. METHODS: Orbital adipose/connective tissue specimens were obtained from euthyroid patients undergoing orbital decompression surgery for severe GO (n = 9) and from normal individuals (n = 9). Thy-1 mRNA and protein levels were assessed in tissue specimens and in orbital fibroblast cultures at baseline using RT-PCR, quantitative immunofluorescent staining, and flow cytometry using a specific Thy-1 mouse anti-human CD90/Thy-1 monoclonal antibody. In addition, some orbital fibroblast cultures were treated with rosiglitazone (1 microL/mL; 2 nM) or control for 10 days in culture. RESULTS: We found that Thy-1 mRNA and protein expression was higher in uncultured GO connective/adipose tissue specimens (3.8-fold; 0.835 +/- 0.116 relative expression) compared with normal (0.22 +/- 0.062; p = 0.002) and in cultured orbital fibroblasts from GO patients (3.3-fold; 9.28 +/- 1.82 relative expression) compared with normal cultures (2.80 +/- 0.42; p = 0.013). Adipocyte differentiation had no effect on Thy-1 expression. Flow cytometry and immunofluorescent staining showed increased numbers of Thy-1-positive cells in the GO (mean 77.9 + 4.09%; range 66.5-84.8%) compared with the normal fibroblast cultures (66.8 + 1.6%; range 63.3-71.0% positive; p = 0.046), as well as higher levels of expression on the positive cells. CONCLUSIONS: Increased Thy-1 expression in GO orbital tissues and cultures is likely a consequence of the orbital disease process, reflecting both the presence of increased numbers of Thy-1-positive cells and higher expression on those cells. Adipogenesis itself does not appear to impact Thy-1 expression. Increased expression of this protein in GO could represent an adaptive response to cell injury, in effect limiting disease progression within the orbital adipose/connective tissues.


Assuntos
Oftalmopatia de Graves/metabolismo , Órbita/metabolismo , Antígenos Thy-1/biossíntese , Tecido Adiposo/metabolismo , Adulto , Animais , Tecido Conjuntivo/metabolismo , Feminino , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo
17.
Thyroid ; 15(5): 422-6, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15929662

RESUMO

Thyroid transcription factor-1 (TTF-1) is required for maximal expression of thyrotropin receptor (TSHR) in the thyroid. Extrathyroidal TSHR expression is detectable in normal orbital adipose tissues, with increased levels found in orbital tissues from patients with Graves' ophthalmopathy (GO), and in orbital preadipocyte cultures following differentiation. In order to determine whether TTF-1 might be involved in orbital TSHR expression, we used quantitative real-time polymerase chain reaction (PCR) to assess relative expression of this and other thyroid-associated transcription factors (TTF-2 and Pax-8) in GO orbital tissue specimens (n = 28) and cultures (n = 3), and in normal orbital tissues (n = 19) and cultures (n = 3). We detected TTF-1 and TTF-2 mRNA in GO and normal orbital tissue samples, with no difference in levels noted between the tissues. In the GO orbital cultures, TTF-1 mRNA was higher in differentiated than in control (undifferentiated) cultures (p < 0.05), while TTF-2 was unchanged. In the normal cultures, neither TTF-1 nor TTF-2 mRNA levels increased in differentiated cultures. Pax8 was undetectable in all orbital tissues and cell cultures. The presence of mRNA encoding TTF-1 in orbital tissues and cultures suggest that this transcription factor may play an important role in extrathyroidal, as it does in thyroidal, TSHR expression.


Assuntos
Tecido Adiposo/metabolismo , Proteínas Nucleares/metabolismo , Órbita/metabolismo , Receptores da Tireotropina/biossíntese , Fatores de Transcrição/metabolismo , Adenosina Trifosfatases , Adipócitos/metabolismo , Diferenciação Celular , Células Cultivadas , Primers do DNA/farmacologia , DNA Complementar/biossíntese , DNA Complementar/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Fator de Transcrição PAX8 , Fatores de Transcrição Box Pareados , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Nuclear 1 de Tireoide , Transativadores/metabolismo
18.
J Clin Endocrinol Metab ; 90(8): 4730-5, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15886250

RESUMO

CONTEXT: The signs and symptoms of Graves' ophthalmopathy (GO) result from inflammation and increased volume of the orbital adipose tissues and extraocular muscles. OBJECTIVE: Our objective was to identify differentially regulated genes that may be involved in stimulating the orbital adipose tissue expansion seen in GO. DESIGN: Gene expression profiling was used to compare genes expressed in orbital adipose tissues from GO patients and normal individuals. SETTING: The study took place at a private practice tertiary referral center. PATIENTS: Orbital adipose tissues were collected at transantral orbital decompression surgery from 20 euthyroid patients undergoing this procedure for severe GO and at early autopsy from eight normal individuals having no evidence of thyroid or ocular disease. RESULTS: Of the 12,686 genes analyzed, 25 known genes were increased in expression (>4-fold) in GO orbital tissues, whereas 11 genes were decreased (>4-fold). Up-regulated genes, confirmed by quantitative RT-PCR, included secreted frizzled-related protein-1 (sFRP-1; 18.5-fold) and several adipocyte-related genes, including peroxisome proliferator activated receptor-gamma (44.1-fold) and adiponectin (25-fold). Treatment in vitro of GO orbital preadipocytes with recombinant sFRP-1 (100 nm) significantly increased adiponectin (2.0-fold; P < 0.05), leptin (7-fold; P < 0.002), and TSH receptor mRNA (13-fold; P < 0.003) levels and enhanced Oil red-O staining in the cultures. CONCLUSIONS: These results support the concept that orbital adipogenesis is enhanced in GO and suggest that elevated levels of sFRP-1 in the GO orbit may be involved in stimulating this pathogenic process.


Assuntos
Tecido Adiposo/fisiologia , Perfilação da Expressão Gênica , Doença de Graves/genética , Doença de Graves/fisiopatologia , Proteínas/genética , Adipócitos/citologia , Adipócitos/fisiologia , Tecido Adiposo/citologia , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Órbita , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
19.
J Clin Endocrinol Metab ; 89(2): 930-5, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14764816

RESUMO

The signs and symptoms of Graves' ophthalmopathy (GO) result from increased volume of the orbital contents, including adipose, connective, and extraocular muscle tissues. We wanted to determine whether the expanded adipose tissue volume might be in part attributable to de novo adipogenesis. We measured levels of mRNA encoding leptin, adiponectin, peroxisome proliferator-activated receptor gamma (PPAR gamma), preadipocyte factor-1, and TSH receptor (TSHr) genes in orbital adipose tissues from GO patients (n = 22) and normal individuals (n = 18) and in orbital preadipocyte cultures derived from GO patients (n = 6) and normal subjects (n = 3) using quantitative real-time RT PCR. We found increased leptin, adiponectin, PPAR gamma, and TSHr expression in GO compared with normal orbital tissue samples, with positive correlations in the GO tissues between TSHr and leptin, adiponectin and PPAR gamma. In vitro differentiation of GO and normal preadipocytes resulted in enhanced adiponectin, leptin, and TSHr expression, with greater expression of the latter two genes in the GO cultures. These results suggest that de novo adipogenesis within orbital tissues with parallel enhanced expression of TSHr may be important in the pathogenesis of GO, and that potential therapies for GO might include inhibition of the adipogenic pathway.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Doença de Graves/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Órbita/crescimento & desenvolvimento , Adipócitos/patologia , Adiponectina , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Estudos de Casos e Controles , Diferenciação Celular , Células Cultivadas , Doença de Graves/patologia , Humanos , Leptina/genética , Órbita/metabolismo , Órbita/patologia , Proteínas/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores da Tireotropina/genética , Células-Tronco/patologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA