Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 660
Filtrar
1.
Fish Shellfish Immunol ; 134: 108593, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36746229

RESUMO

The inhibition of inflammatory response is an essential process to control the development of inflammation and is an important step to protect the organism from excessive inflammatory damage. As a pleiotropic cytokine, transforming growth factor beta (TGF-ß) plays a regulatory role in inhibiting inflammation in vertebrates. To investigate the role of TGF-ß in the regulation of inflammation in invertebrates, we cloned and characterized the TGF-ß gene from Apostichopus japonicus via rapid amplification of cDNA ends, and the sample was designated as AjTGF-ß. For Vibrio splendidus-challenged sea cucumbers, the expression of AjTGF-ß mRNAs in coelomocytes decreased at 96 h (0.27-fold), which was contrary to the trend of inflammation. AjTGF-ß was expressed in all tissues with the highest expression in the body wall. When AjTGF-ß was knocked down by using small interfering RNA (siRNA-KD) to 0.45-fold, AjSMAD 2/3 and AjSMAD6 were downregulated to 0.32- and 0.05-fold compared with the control group, respectively. Furthermore, when the damaged sea cucumber was challenged by V. splendidus co-incubated with rAjTGF-ß, the damage area had no extensive inflammation, and damaged repair appeared at 72 h compared with the Vs + BSA group, in which the expression of AjSMAD 2/3 was upregulated by 1.35-fold. Under this condition, AjSMAD 2/3 silencing alleviated rAjTGF-ß-induced damage recovery. Moreover, rAjTGF-ß slightly induced the collagen I expression from 6.13 ng/mL to 7.84 ng/mL, and collagen III was upregulated from 6.23 ng/mL to 6.89 ng/mL compared with the Vs + BSA group. This finding indicates that AjTGF-ß negatively regulated the inflammatory progress and accelerated the repair of damage by AjSMADs to regulate the collagens expression.


Assuntos
Proteínas Smad , Stichopus , Fator de Crescimento Transformador beta , Sequência de Aminoácidos , Invertebrados/classificação , Invertebrados/genética , Invertebrados/imunologia , Modelos Moleculares , Filogenia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Proteínas Smad/metabolismo , Stichopus/classificação , Stichopus/genética , Stichopus/imunologia , Stichopus/microbiologia , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Animais
2.
Biochim Biophys Acta Proteins Proteom ; 1870(3): 140746, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34942360

RESUMO

Mutations in the fasciclin 1 domain 4 (FAS1-4) of transforming growth factor ß-induced protein (TGFBIp) are associated with insoluble extracellular deposits and corneal dystrophies (CDs). The decrease in solubility upon mutation has been implicated in CD; however, the exact molecular mechanisms are not well understood. Here, we performed molecular dynamics simulations followed by solvation thermodynamic analyses of the FAS1-4 domain and its three mutants-R555W, R555Q, and A546T-linked to granular corneal dystrophy type 1, Thiel-Behnke corneal dystrophy and lattice corneal dystrophy, respectively. We found that both R555W and R555Q mutants have less affinity toward solvent water relative to the wild-type protein. In the R555W mutant, a remarkable increase in solvation free energy was observed because of the structural changes near the mutation site. The mutation site W555 is buried in other hydrophobic residues, and R557 simultaneously forms salt bridges with E554 and D561. In the R555Q mutant, the increase in solvation free energy is caused by structural rearrangements far from the mutation site. R558 separately forms salt bridges with D575, E576, and E598. Thus, we thus identified the relationship between the decrease in solubility and conformational changes caused by mutations, which may be useful in designing potential therapeutics and in blocking FAS1 aggregation related to CD.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Distrofias Hereditárias da Córnea/genética , Proteínas da Matriz Extracelular/genética , Mutação , Fator de Crescimento Transformador beta/genética , Amiloide/química , Amiloide/metabolismo , Moléculas de Adesão Celular Neuronais/química , Distrofias Hereditárias da Córnea/metabolismo , Proteínas da Matriz Extracelular/química , Humanos , Simulação de Dinâmica Molecular , Estrutura Molecular , Agregação Patológica de Proteínas/metabolismo , Solubilidade , Fator de Crescimento Transformador beta/química
3.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34681785

RESUMO

The aim of this study was to compare the bone regeneration ability of particle and block bones, acting as bone scaffolds, with recombinant human bone morphogenetic protein (rhBMP)-2 and evaluate them as rhBMP-2 carriers. Demineralized bovine bone particles, blocks, and rhBMP-2 were grafted into the subperiosteal space of a rat calvarial bone, and the rats were randomly divided into four groups: particle, block, P (particle)+BMP, and B (block)+BMP groups. The bone volume of the B+BMP group was significantly higher than that of the other groups (p < 0.00), with no significant difference in bone mineral density. The average adipose tissue volume of the B+BMP group was higher than that of the P+BMP group, although the difference was not significant. Adipose tissue formation was observed in the rhBMP-2 application group. Histologically, the particle and B+BMP groups showed higher formation of a new bone. However, adipose tissue and void spaces were also formed, especially in the B+BMP group. Hence, despite the formation of a large central void space, rhBMP-2 could be effectively used with block bone scaffolds and showed excellent new bone formation. Further studies are required to evaluate the changes in adipose tissue.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea/efeitos dos fármacos , Alicerces Teciduais/química , Fator de Crescimento Transformador beta/farmacologia , Animais , Proteína Morfogenética Óssea 2/química , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Bovinos , Técnicas de Cultura de Células/métodos , Meios de Cultura/química , Meios de Cultura/farmacologia , Humanos , Osteogênese/efeitos dos fármacos , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Crânio , Fator de Crescimento Transformador beta/química
4.
Carbohydr Polym ; 273: 118589, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34560990

RESUMO

Nowadays, vascularization and mineralization of bone defects is the main bottleneck in the bone regeneration field that is needed to be overcome and developed. Here, we prepared novel in-situ formed injectable hydrogels based on chitosan biguanidine and carboxymethylcellulose loaded with vascular endothelial growth factor (VEGF) and recombinant Bone morphogenetic protein 2 (BMP-2) and studied its influence on osteoblastic differentiation of dental pulp stem cells (DPSCs). The sequential release behavior of the VEGF and BMP-2 from hydrogels adjusted with the pattern of normal human bone growth. MTT assay exhibited that these hydrogels were non-toxic and significantly increased DPSCs proliferation. The Real-time PCR and Western blot analysis on CG11/BMP2-VEGF showed significantly higher gene and protein expression of ALP, COL1α1, and OCN. These results were confirmed by mineralization assay by Alizarin Red staining and Alkaline phosphatase enzyme activity. Based on these evaluations, these hydrogel holds potential as an injectable bone tissue engineering platform.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Portadores de Fármacos/química , Hidrogéis/química , Osteogênese/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteína Morfogenética Óssea 2/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quitosana/análogos & derivados , Quitosana/toxicidade , Polpa Dentária/citologia , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Guanidinas/química , Guanidinas/toxicidade , Humanos , Hidrogéis/toxicidade , Osteoblastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Resistência à Tração , Alicerces Teciduais/química , Fator de Crescimento Transformador beta/química , Fator A de Crescimento do Endotélio Vascular/química
5.
J Mater Sci Mater Med ; 32(9): 107, 2021 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-34427778

RESUMO

OBJECTIVE: To study the bone induction and defect repair of true bone ceramics (TBC) combined with rhBMP-2 and Sr. METHODS: MC3T3-E1 cells were used to evaluate the bioactivity of the composite. Cell proliferation activity was detected by CCK-8, ALP activity was detected by p-nitrophenyl phosphate (PNPP), and the differences of material surface topography were observed by scanning electron microscopy (SEM). Bone induction was verified by the implantation in nude mice. The rabbit femoral condyle defect model was achieved to verify the bone defect repair ability of the material. RESULTS: SEM results showed nearly the same surface morphology and cell proliferation quantified by CCK-8 showed that compared with TBC, both TBC&Sr and TBC&BMP-2&Sr had a significant promoting effect (P < 0.05). ALP activity result showed that the ALP activity of TBC&BMP-2&Sr was significantly higher than that of TBC alone (P < 0.05). The bone induction result showed that TBC&Sr had a small amount of new bone formation, and the new bone area was only 2.5 ± 0.11%. The bone induction activity of TBC&BMP-2&Sr was the highest, the new bone area was up to 75.36 ± 4.21%. Histological result of bone defect repair showed that TBC&BMP-2&Sr was also the highest, the new bone area was up to 72.42 ± 3.14%. The repair effect of TBC& BMP-2 was second, and better than that of TBC&Sr. CONCLUSION: TBC combined with rhBMP-2 and Sr had the good bioactivity, obvious bone conduction and bone defect repair performance, laying the foundation of clinical application potentially.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Estrôncio/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Proteína Morfogenética Óssea 2/química , Substitutos Ósseos/química , Substitutos Ósseos/farmacologia , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cerâmica/química , Cerâmica/farmacologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Feminino , Fraturas Ósseas/terapia , Masculino , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Estrôncio/química , Alicerces Teciduais/química , Fator de Crescimento Transformador beta/química
6.
Biomed Mater ; 16(5)2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34181586

RESUMO

The onlay-graft, one of the most difficult graft conditions, is used for diverse clinical conditions, including plastic and dental surgery. The graft should withstand continuous pressure from overlying tissues and have excellent bone formation capability in a limited bone contact situation. We recently developed a 3D printed Kagome-structured polycaprolactone (PCL) scaffold that has a stronger mechanical property. This study evaluated the clinical feasibility of this scaffold for onlay-graft use. The value of the scaffold containing recombinant human bone morphogenetic protein-2 in a hyaluronate-based hydrogel (rhBMP-2/HA) to enhance bone regeneration was also assessed. 3D-printed Kagome-PCL scaffolds alone (n= 12, group I) or loaded with rhBMP-2/HA (n= 12, group II) were grafted using a rat calvarial onlay-graft model. Following sacrifice at 2, 4, and 8 weeks, all 3D-printed Kagome-PCL scaffolds were accurately positioned and firmly integrated to the recipient bone. Micro-computed tomography and histology analyses revealed a constant height of the scaffolds over time in all animals. New bone grew into the scaffolds in both groups, but with greater volume in group II. These results suggest the promising clinical feasibility of the 3D-printed Kagome-PCL scaffold for onlay-graft use and it could substitute the conventional onlay-graft in the plastic and dental reconstructive surgery in the near future.


Assuntos
Proteína Morfogenética Óssea 2 , Ácido Hialurônico , Poliésteres , Impressão Tridimensional , Alicerces Teciduais/química , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea/efeitos dos fármacos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Masculino , Poliésteres/química , Poliésteres/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/farmacologia
7.
Front Immunol ; 12: 628059, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122401

RESUMO

Artificial antigen-presenting cells (aAPCs) are synthetic versions of naturally occurring antigen-presenting cells (APCs) that, similar to natural APCs, promote efficient T effector cell responses in vitro. This report describes a method to produce acellular tolerogenic aAPCs made of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) and encapsulating IL-2 and TGF-ß for a paracrine release to T cells. We document that these aAPCs can induce both human CD4+ and CD8+ T cells to become FoxP3+ T regulatory cells (Tregs). The aAPC NP-expanded human Tregs are functional in vitro and can modulate systemic autoimmunity in vivo in humanized NSG mice. These findings establish a proof-of-concept to use PLGA NPs as aAPCs for the induction of human Tregs in vitro and in vivo, highlighting the immunotherapeutic potential of this targeted approach to repair IL-2 and/or TGF-ß defects documented in certain autoimmune diseases such as systemic lupus erythematosus.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Doença Enxerto-Hospedeiro/prevenção & controle , Interleucina-2/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/transplante , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Humanos , Interleucina-2/química , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Estudo de Prova de Conceito , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/química
8.
Food Funct ; 12(11): 5038-5050, 2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-33960359

RESUMO

Rheumatoid Arthritis (RA) is an autoimmune disease that commences as inflammation and progressively destroys the articular joint. In this study, we assess the anti-rheumatic potential of the monoterpenoid class of thymol conjugated with Carbon Dots (CDs). Waste biomass in the form of dried rose petals was chosen as a precursor for the synthesis of CDs via a one-step hydrothermal bottom-up methodology. The prepared CDs exhibited absorption in the near-visible region, and unique excitation-dependent emission behaviour was confirmed from UV-Visible and fluorescence measurements. The surface morphology of CDs was confirmed by SEM and HR-TEM analysis to be quasi-spherical particles with an average size of ∼5-6 nm. The presence of various functional moieties (hydroxyl, carbonyl, and amino) was confirmed via FT-IR measurement. The graphitization of CDs was confirmed by the D and G bands for sp2 and sp3 hybridization, respectively, through Raman analysis. Esterification methodology was adopted to prepare the CDs-thymol conjugate and confirmed via FT-IR analysis. CDs play the role of a nanocarrier for thymol, an anti-arthritic agent. The bioactive compound of thymol showed potent anti-arthritic activity against RA targets through in silico docking studies. Further, the in vivo studies revealed that CDs-thymol conjugates (10 mg per kg body weight) showed a significant reduction in rat paw volume along with reduced levels of RF and CRP (2.23 ± 0.42 IU ml-1 and 16.96 ± 0.22 mg ml-1) when compared to the disease control rats. X-ray radiography and ultrasonic imaging revealed less bone destruction, joint derangement, and swelling in arthritis-induced Wistar rats. They could also potentially improve the Hb (14.14 ± 0.19), RBC (6.01 ± 0.11), PCV (6.01 ± 0.11) levels and elevate the status of antioxidant enzymes (GPx, SOD, MDA), and the activity was comparable to the standard drug, ibuprofen (10 mg kg-1), suggesting that the CDs-thymol conjugate at 10 mg kg-1 could act as a strong anti-arthritic agent. This work is evidence for the utilization of waste biomass as a value-added product such as a nanocarrier for biomedical applications.


Assuntos
Antirreumáticos/química , Antirreumáticos/farmacologia , Carbono/química , Pontos Quânticos , Timol/química , Animais , Antioxidantes , Artrite Reumatoide/tratamento farmacológico , Feminino , Interleucinas/química , Metaloproteinase 1 da Matriz/química , Metaloproteinase 3 da Matriz/química , Simulação de Acoplamento Molecular , Ratos , Ratos Wistar , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Espectroscopia de Infravermelho com Transformada de Fourier , Fator de Crescimento Transformador beta/química
9.
Front Immunol ; 12: 613438, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054795

RESUMO

Transforming growth factor-ß (TGF-ß) isoforms are secreted as inactive complexes formed through non-covalent interactions between bioactive TGF-ß entities and their N-terminal pro-domains called latency-associated peptides (LAP). Extracellular activation of latent TGF-ß within this complex is a crucial step in the regulation of TGF-ß activity for tissue homeostasis and immune cell function. We previously showed that the matrix glycoprotein Tenascin-X (TN-X) interacted with the small latent TGF-ß complex and triggered the activation of the latent cytokine into a bioactive TGF-ß. This activation most likely occurs through a conformational change within the latent TGF-ß complex and requires the C-terminal fibrinogen-like (FBG) domain of the glycoprotein. As the FBG-like domain is highly conserved among the Tenascin family members, we hypothesized that Tenascin-C (TN-C), Tenascin-R (TN-R) and Tenascin-W (TN-W) might share with TN-X the ability to regulate TGF-ß bioavailability through their C-terminal domain. Here, we demonstrate that purified recombinant full-length Tenascins associate with the small latent TGF-ß complex through their FBG-like domains. This association promotes activation of the latent cytokine and subsequent TGF-ß cell responses in mammary epithelial cells, such as cytostasis and epithelial-to-mesenchymal transition (EMT). Considering the pleiotropic role of TGF-ß in numerous physiological and pathological contexts, our data indicate a novel common function for the Tenascin family in the regulation of tissue homeostasis under healthy and pathological conditions.


Assuntos
Tenascina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Homeostase , Humanos , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Proteínas Smad/química , Proteínas Smad/metabolismo , Relação Estrutura-Atividade , Tenascina/química , Tenascina/genética , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética
10.
Biomech Model Mechanobiol ; 20(4): 1627-1644, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34047890

RESUMO

Critical-sized bone defects are critical healing conditions that, if left untreated, often lead to non-unions. To reduce the risk, critical-sized bone defects are often treated with recombinant human BMP-2. Although enhanced bone tissue formation is observed when BMP-2 is administered locally to the defect, spatial and temporal distribution of callus tissue often differs from that found during regular bone healing or in defects treated differently. How this altered tissue patterning due to BMP-2 treatment is linked to mechano-biological principles at the cellular scale remains largely unknown. In this study, the mechano-biological regulation of BMP-2-treated critical-sized bone defect healing was investigated using a multiphysics multiscale in silico approach. Finite element and agent-based modeling techniques were combined to simulate healing within a critical-sized bone defect (5 mm) in a rat femur. Computer model predictions were compared to in vivo microCT data outcome of bone tissue patterning at 2, 4, and 6 weeks postoperation. In vivo, BMP-2 treatment led to complete healing through periosteal bone bridging already after 2 weeks postoperation. Computer model simulations showed that the BMP-2 specific tissue patterning can be explained by the migration of mesenchymal stromal cells to regions with a specific concentration of BMP-2 (chemotaxis). This study shows how computational modeling can help us to further understand the mechanisms behind treatment effects on compromised healing conditions as well as to optimize future treatment strategies.


Assuntos
Proteína Morfogenética Óssea 2/química , Regeneração Óssea/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Colágeno/química , Osteogênese/efeitos dos fármacos , Fator de Crescimento Transformador beta/química , Cicatrização/fisiologia , Animais , Calo Ósseo , Diferenciação Celular , Simulação por Computador , Fêmur/efeitos dos fármacos , Análise de Elementos Finitos , Humanos , Técnicas In Vitro , Células-Tronco Mesenquimais/metabolismo , Ratos , Proteínas Recombinantes/química , Risco , Microtomografia por Raio-X
11.
J Mater Sci Mater Med ; 32(4): 32, 2021 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-33751250

RESUMO

Bone morphogenetic protein two (BMP-2) has been widely used as an osteoinductive agent in the treatment of bone diseases. However, some side effects, such as osteoclast activation have emerged when it was used at high doses. In this study, by considering the osteoclast-suppressing capability of melatonin (MEL), its effect on osteoclast differentiation induced by BMP-2 was investigated. These two factors, MEL and BMP-2, were embedded into chitosan/hydroxyapatite (HAp) scaffolds that were characterized morphologically by scanning electron microscopy (SEM) and micro-computed tomography (µ-CT). Release profiles of MEL and BMP-2 from scaffolds were determined in vitro and then, the differentiation of RAW 264.7 cells to osteoclasts was investigated on the scaffolds. Results of tartrate-resistant acid phosphatase (TRAP) staining, SEM imaging and expression of cathepsin K gene showed that, in the presence of BMP-2, osteoclast differentiation increased, whereas it decreased in MEL and MEL/BMP-2 embedded scaffolds suggesting that melatonin successfully attenuated osteoclast differentiation induced by BMP-2. Thus, the MEL/BMP-2 loaded chitosan/HAp scaffolds that have dual function in enhancing bone formation and inhibiting osteoclast activity are recommended biomaterials in the field of bone regeneration.


Assuntos
Proteína Morfogenética Óssea 2/química , Sistemas de Liberação de Medicamentos , Melatonina/química , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Alicerces Teciduais/química , Fator de Crescimento Transformador beta/química , Animais , Materiais Biocompatíveis , Regeneração Óssea , Reabsorção Óssea/tratamento farmacológico , Catepsina K/biossíntese , Diferenciação Celular , Sobrevivência Celular , Quitosana/química , Durapatita/química , Técnicas In Vitro , Camundongos , Microscopia Eletrônica de Varredura , Células RAW 264.7 , Proteínas Recombinantes/química , Estresse Mecânico , Termogravimetria , Microtomografia por Raio-X
12.
Development ; 148(5)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33712443

RESUMO

The transforming growth factor ß (TGFß) signaling family is evolutionarily conserved in metazoans. The signal transduction mechanisms of TGFß family members have been expansively investigated and are well understood. During development and homeostasis, numerous TGFß family members are expressed in various cell types with temporally changing levels, playing diverse roles in embryonic development, adult tissue homeostasis and human diseases by regulating cell proliferation, differentiation, adhesion, migration and apoptosis. Here, we discuss the molecular mechanisms underlying signal transduction and regulation of the TGFß subfamily pathways, and then highlight their key functions in mesendoderm induction, dorsoventral patterning and laterality development, as well as in the formation of several representative tissues/organs.


Assuntos
Desenvolvimento Embrionário/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Camadas Germinativas/metabolismo , Proteína Nodal/metabolismo , Organogênese , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/química
13.
J Biomed Mater Res A ; 109(6): 840-848, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32776655

RESUMO

Recombinant human bone morphogenetic protein 2 (rhBMP-2) has been widely used in bone tissue engineering to enhance bone regeneration because of its osteogenic inductivity. However, clinical outcomes can vary depending on the scaffold materials used to deliver rhBMP-2. In this study, 3D-printed scaffolds with a ratio of 1:1 polycaprolactone and beta-tricalcium phosphate (PCL/T50) were applied as carriers for rhBMP-2 in mandibular bone defect models in dog models. Before in vivo application, in vitro experiments were conducted. Preosteoblast proliferation was not significantly different between scaffolds made of PCL/T50 and polycaprolactone alone (PCL/T0) regardless of rhBMP-2 delivery. However, PCL/T50 showed an increased level of the alkaline phosphatase activity and mineralization assay when rhBMP-2 was delivered. In in vivo, the newly formed bone volume of the PCL/T50 group was significantly increased compared with that of the PCL/T0 scaffolds regardless of rhBMP-2 delivery. Histological examination showed that PCL/T50 with rhBMP-2 produced significantly greater amounts of newly bone formation than PCL/T0 with rhBMP-2. The quantities of scaffold remaining were lower in the PCL/T50 group than in the PCL/T0 group, although it was not significantly different. In conclusion, PCL/T50 scaffolds were advantageous for rhBMP-2 delivery as well as for maintaining space for bone formation in mandibular bone defects.


Assuntos
Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Fosfatos de Cálcio/química , Osteogênese/efeitos dos fármacos , Poliésteres/química , Alicerces Teciduais , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/farmacologia , Fosfatase Alcalina/química , Animais , Materiais Biocompatíveis , Regeneração Óssea , Proliferação de Células/efeitos dos fármacos , Cães , Portadores de Fármacos , Masculino , Mandíbula/anormalidades , Impressão Tridimensional , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Células-Tronco
14.
Sci Signal ; 13(662)2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33323411

RESUMO

Transforming growth factor-ß (TGF-ß) proteins regulate multiple cellular functions, including cell proliferation, apoptosis, and extracellular matrix formation. The dysregulation of TGF-ß signaling causes diseases such as cancer and fibrosis, and therefore, understanding the biochemical basis of TGF-ß signal transduction is important for elucidating pathogenic mechanisms in these diseases. SMAD proteins are transcription factors that mediate TGF-ß signaling-dependent gene expression. The transcriptional coactivator CBP directly interacts with the MH2 domains of SMAD2 to activate SMAD complex-dependent gene expression. Here, we report the structural basis for CBP recognition by SMAD2. The crystal structures of the SMAD2 MH2 domain in complex with the SMAD2-binding region of CBP showed that CBP forms an amphiphilic helix on the hydrophobic surface of SMAD2. The expression of a mutated CBP peptide that showed increased SMAD2 binding repressed SMAD2-dependent gene expression in response to TGF-ß signaling in cultured cells. Disrupting the interaction between SMAD2 and CBP may therefore be a promising strategy for suppressing SMAD-dependent gene expression.


Assuntos
Fragmentos de Peptídeos/química , Sialoglicoproteínas/química , Transdução de Sinais , Proteína Smad2/química , Fator de Crescimento Transformador beta/química , Humanos , Fragmentos de Peptídeos/metabolismo , Domínios Proteicos , Sialoglicoproteínas/metabolismo , Proteína Smad2/metabolismo , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta/metabolismo
15.
PLoS One ; 15(9): e0239396, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32966314

RESUMO

Despite recent progress in the treatment of rheumatoid arthritis (RA), many patients still fail to achieve remission or low disease activity. An imbalance between auto-reactive effector T cells (Teff) and regulatory T cells (Treg) may contribute to joint inflammation and damage in RA. Therefore, restoring this balance is a promising approach for the treatment of inflammatory arthritis. Accordingly, our group has previously shown that the combination of TGF-ß-releasing microparticles (MP), rapamycin-releasing MP, and IL-2-releasing MP (TRI MP) can effectively increase the ratio of Tregs to Teff in vivo and provide disease protection in several preclinical models. In this study TRI MP was evaluated in the collagen-induced arthritis (CIA) model. Although this formulation has been tested previously in models of destructive inflammation and transplantation, this is the first model of autoimmunity for which this therapy has been applied. In this context, TRI MP effectively reduced arthritis incidence, the severity of arthritis scores, and bone erosion. The proposed mechanism of action includes not only reducing CD4+ T cell proliferation, but also expanding a regulatory population in the periphery soon after TRI MP administration. These changes were reflected in the CD4+ T cell population that infiltrated the paws at the onset of arthritis and were associated with a reduction of immune infiltrate and inflammatory myeloid cells in the paws. TRI MP administration also reduced the titer of collagen antibodies, however the contribution of this reduced titer to disease protection remains uncertain since there was no correlation between collagen antibody titer and arthritis score.


Assuntos
Artrite Experimental/prevenção & controle , Interleucina-2/farmacologia , Microesferas , Sirolimo/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Autoanticorpos/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Contagem de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Interleucina-2/química , Masculino , Camundongos , Sirolimo/química , Fator de Crescimento Transformador beta/química
17.
Mater Sci Eng C Mater Biol Appl ; 111: 110750, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32279822

RESUMO

A promising direction for the replacement of expanded bone defects is the development of bioimplants based on synthetic biocompatible materials impregnated with growth factors that stimulate bone remodeling. Novel biomimetic highly porous ultra-high molecular weight polyethylene (UHMWPE)/40% hydroxyapatite (HA) scaffold for reconstructive surgery with the porosity of 85 ± 1% vol. and a diameter of pores in the range of 50-800 µm was developed. The manufacturing process allowed the formation of trabecular-like architecture without additional solvents and thermo-oxidative degradation. Biomimetic UHMWPE/HA scaffold was biocompatible and provided effective tissue ingrowth on a model of critical-sized cranial defects in mice. The combined use of UHMWPE/HA with Bone Morphogenetic Protein-2 (BMP-2) demonstrated intensive mineralized bone formation as early as 3 weeks after surgery. The addition of erythropoietin (EPO) significantly enhanced angiogenesis in newly formed tissues. The effect of EPO of bacterial origin on bone tissue defect healing was demonstrated for the first time. The developed biomimetic highly porous UHMWPE/HA scaffold can be used separately or in combination with rhBMP-2 and EPO for reconstructive surgery to solve the problems associated with difference between implant architecture and trabecular bone, low osteointegration and bioinertness.


Assuntos
Materiais Biocompatíveis/química , Doenças Ósseas/cirurgia , Proteína Morfogenética Óssea 2/química , Durapatita/química , Eritropoetina/química , Polietilenos/química , Fator de Crescimento Transformador beta/química , Animais , Materiais Biocompatíveis/farmacologia , Doenças Ósseas/terapia , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/uso terapêutico , Regeneração Óssea/efeitos dos fármacos , Osso e Ossos/patologia , Osso e Ossos/fisiologia , Portadores de Fármacos/química , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Nanocompostos/química , Neovascularização Fisiológica/efeitos dos fármacos , Porosidade , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Procedimentos de Cirurgia Plástica , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/uso terapêutico
18.
J Mater Chem B ; 8(17): 3842-3851, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32219244

RESUMO

Developing a biomaterial that can promote osteoblastic differentiation, thereby reducing the needs of exogenous osteogenic factors for large bone repair, has been a significant and long-term technical hurdle. In this study, we developed an innovative nanoclay (nanosilicate, NS)-functionalized 3D gelatin nanofibrous scaffold (GF/NS) through a thermally induced phase separation method together with the particle leaching technique (TIPS&P). In addition to the significantly higher mechanical strength, the composite scaffolds (GF/NS) demonstrated a significantly stronger ability to promote the osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro compared to the GF scaffold. Our data further revealed that this intriguing pro-osteoblastic functionality was largely because of the unique features of NS, particularly, the strong binding ability to pro-osteoblastic factors (e.g., BMP2) as well as the intrinsic osteoinductivity of its bioactive degradation products. Most importantly, our in vivo studies indicated that GF/NS scaffolds significantly improved low-dose BMP2-induced ectopic bone regeneration in mice.


Assuntos
Regeneração Óssea/fisiologia , Nanofibras/química , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/uso terapêutico , Doenças Ósseas/patologia , Doenças Ósseas/terapia , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Módulo de Elasticidade , Gelatina/química , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanofibras/toxicidade , Osteogênese/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Silicatos/química , Engenharia Tecidual , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/metabolismo
19.
Neoplasia ; 22(4): 163-178, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32143140

RESUMO

BUB1 (budding uninhibited by benzimidazoles-1) is required for efficient TGF-ß signaling, through its role in stabilizing the TGFBR1 and TGFBR2 complex. Here we demonstrate that TGFBR2 phosphorylates BUB1 at Serine-318, which is conserved in primates. S318 phosphorylation abrogates the interaction of BUB1 with TGFBR1 and SMAD2. Using BUB1 truncation domains (1-241, 241-482 and 482-723), we demonstrate that multiple contact points exist between BUB1 and TGF-ß signaling components and that these interactions are independent of the BUB1 tetratricopeptide repeat (TPR) domain. Moreover, substitutions in the middle domain (241-482) encompassing S318 reveals that efficient interaction with TGFBR2 occurs only in its dephosphorylated state (241-482 S318A). In contrast, the phospho-mimicking mutant (241-482 S318D) exhibits efficient binding with SMAD2 and its over-expression results in a decrease in TGFBR1-TGFBR2 and TGFBR1-SMAD2 interactions. These findings suggest that TGFBR2 mediated BUB1 phosphorylation at S318 may serve as a switch for the dissociation of the SMAD2-TGFBR complex, and therefore represents a regulatory event for TGF-ß signaling. Finally, we provide evidence that the BUB1-TGF-ß signaling axis may mediate aggressive phenotypes in a variety of cancers.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Serina/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/química , Receptor do Fator de Crescimento Transformador beta Tipo II/química , Fator de Crescimento Transformador beta/química
20.
Sci Rep ; 10(1): 4011, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32132634

RESUMO

Protein aggregation has been one of the leading triggers of various disease conditions, such as Alzheimer's, Parkinson's and other amyloidosis. TGFBI-associated corneal dystrophies are protein aggregation disorders in which the mutant TGFBIp aggregates and accumulates in the cornea, leading to a reduction in visual acuity and blindness in severe cases. Currently, the only therapy available is invasive and there is a known recurrence after surgery. In this study, we tested the inhibitory and amyloid dissociation properties of four osmolytes in an in-vitro TGFBI peptide aggregation model. The 23-amino acid long peptide (TGFBIp 611-633 with the mutation c.623 G>R) from the 4th FAS-1 domain of TGFBIp that rapidly forms amyloid fibrils was used in the study. Several biophysical methods like Thioflavin T (ThT) fluorescence, Circular Dichroism (CD), fluorescence microscopy and Transmission electron microscopy (TEM) were used to study the inhibitory and amyloid disaggregation properties of the four osmolytes (Betaine, Raffinose, Sarcosine, and Taurine). The osmolytes were effective in both inhibiting and disaggregating the amyloid fibrils derived from TGFBIp 611-633 c.623 G>R peptide. The osmolytes did not have an adverse toxic effect on cultured human corneal fibroblast cells and could potentially be a useful therapeutic strategy for patients with TGFBIp corneal dystrophies.


Assuntos
Amiloide , Córnea , Proteínas da Matriz Extracelular , Fibroblastos , Peptídeos , Agregação Patológica de Proteínas , Fator de Crescimento Transformador beta , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Linhagem Celular , Córnea/metabolismo , Córnea/patologia , Distrofias Hereditárias da Córnea/genética , Distrofias Hereditárias da Córnea/metabolismo , Distrofias Hereditárias da Córnea/patologia , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...