Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Wound Repair Regen ; 22(6): 740-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25231003

RESUMO

Fibrin biomatrices have been used for many years for hemostasis and sealing and are a well-established surgical tool. The objective of the present study was to compare two commercially available fibrin biomatrices regarding the effect of their thrombin concentration on keratinocytes and wound healing in vitro and in vivo. Keratinocytes showed significant differences in adhesion, viability, and morphology in the presence of the fibrin matrices in vitro. A high thrombin concentration (800-1,200 IU/mL) caused deteriorated cell compatibility. By using a thrombin inhibitor, those differences could be reversed. In a rat excisional wound healing model, we observed more rapid wound closure and less wound severity in wounds treated with a fibrin matrix containing a lower concentration of thrombin (4 IU/mL). Furthermore, fewer new functional vessels and a lower level of vascular endothelial growth factor were measured in wounds after 7 days treated with the matrix with higher thrombin concentration. These in vivo results may be partially explained by the in vitro biocompatibility data. Additionally, results show that low thrombin biomatrices were degraded faster than the high thrombin material. Hence, we conclude that the composition of fibrin biomatrices influences keratinocytes and therefore has an impact on wound healing.


Assuntos
Materiais Biocompatíveis/farmacologia , Adesivo Tecidual de Fibrina/farmacologia , Pele/efeitos dos fármacos , Trombina/farmacologia , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/tratamento farmacológico , Animais , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Técnicas In Vitro , Queratinócitos , Masculino , Ratos , Ratos Sprague-Dawley , Pele/lesões , Pele/patologia
2.
Cell Tissue Bank ; 15(2): 227-39, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24166477

RESUMO

Human amniotic membrane (hAM) is a tissue containing cells with proven stem cell properties. In its decellularized form it has been successfully applied as nerve conduit biomaterial to improve peripheral nerve regeneration in injury models. We hypothesize that viable hAM without prior cell isolation can be differentiated towards the Schwann cell lineage to generate a possible alternative to commonly applied tissue engineering materials for nerve regeneration. For in vitro Schwann cell differentiation, biopsies of hAM of 8 mm diameter were incubated with a sequential order of neuronal induction and growth factors for 21 days and characterized for cellular viability and the typical glial markers glial fibrillary acidic protein (GFAP), S100ß, p75 and neurotrophic tyrosine kinase receptor (NTRK) using immunohistology. The secretion of the neurotrophic factors brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) was quantified by ELISA. The hAM maintained high viability, especially under differentiation conditions (90.2 % ± 41.6 day 14; 80.0 % ± 44.5 day 21 compared to day 0). Both, BDNF and GDNF secretion was up-regulated upon differentiation. The fresh membrane stained positive for GFAP and p75 and NTRK, which was strongly increased after culture in differentiation conditions. Especially the epithelial layer within the membrane exhibited a change in morphology upon differentiation forming a multi-layered epithelium with intense accumulations of the marker proteins. However, S100ß was expressed at equal levels and equal distribution in fresh and cultured hAM conditions. Viable hAM may be a promising alternative to present formulations used for peripheral nerve regeneration.


Assuntos
Âmnio/citologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Separação Celular , Células de Schwann/citologia , Âmnio/metabolismo , Células Cultivadas , Humanos , Regeneração/fisiologia , Células-Tronco/citologia
3.
Cell Tissue Bank ; 15(2): 213-25, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24828570

RESUMO

Human amniotic membrane (hAM) represents a tissue that is well established as biomaterial in the clinics with potential for new applications in regenerative medicine. For tissue engineering (TE) strategies, cells are usually combined with inductive factors and a carrier substrate. We have previously recognized that hAM represents a natural, preformed sheet including highly potent stem cells. In the present approach for cartilage regeneration we have induced chondrogenesis in hAM in vitro. For this, hAM biopsies were cultured for up to 56 days under chondrogenic conditions. The induced hAM was characterized for remaining viability, glycosaminoglycan (GAG) accumulation using histochemical analysis, and a quantitative assay. Collagen I, II and X was immunohistochemically determined and cartilage-specific mRNA expression of (sex determining region Y-) box 9, cartilage oligomeric matrix protein (COMP), aggrecan (AGC1), versican (CSPG2), COL1A1, COL9A2, melanoma inhibitory activity (MIA), and cartilage-linking protein 1 (CRTL1) analyzed by quantitative real-time polymerase chain reaction. Human AM was successfully induced to accumulate GAG, as demonstrated by Alcianblue staining and a significant (p < 0.001) increase of GAG/viability under chondrogenic conditions peaking in a 29.9 ± 0.9-fold induction on day 56. Further, upon chondrogenic induction collagen II positive areas were identified within histological sections and cartilage-specific markers including COMP, AGC1, CSPG2, COL1A1, COL9A2, MIA, and CRTL1 were found upregulated at mRNA level. This is the first study, demonstrating that upon in vitro induction viable human amnion expresses cartilage-specific markers and accumulates GAGs within the biomatrix. This is a promising first step towards a potential use of living hAM for cartilage TE.


Assuntos
Âmnio/citologia , Diferenciação Celular , Linhagem da Célula/fisiologia , Condrogênese/fisiologia , Placenta/citologia , Cartilagem/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Gravidez , Células-Tronco/citologia
4.
Biomolecules ; 14(4)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38672430

RESUMO

Bovine serum albumin (BSA) plays a crucial role in cell culture media, influencing cellular processes such as proliferation and differentiation. Although it is commonly included in chondrogenic differentiation media, its specific function remains unclear. This study explores the effect of different BSA concentrations on the chondrogenic differentiation of human adipose-derived stromal/stem cells (hASCs). hASC pellets from six donors were cultured under chondrogenic conditions with three BSA concentrations. Surprisingly, a lower BSA concentration led to enhanced chondrogenesis. The degree of this effect was donor-dependent, classifying them into two groups: (1) high responders, forming at least 35% larger, differentiated pellets with low BSA in comparison to high BSA; (2) low responders, which benefitted only slightly from low BSA doses with a decrease in pellet size and marginal differentiation, indicative of low intrinsic differentiation potential. In all cases, increased chondrogenesis was accompanied by hypertrophy under low BSA concentrations. To the best of our knowledge, this is the first study showing improved chondrogenicity and the tendency for hypertrophy with low BSA concentration compared to standard levels. Once the tendency for hypertrophy is understood, the determination of BSA concentration might be used to tune hASC chondrogenic or osteogenic differentiation.


Assuntos
Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais , Soroalbumina Bovina , Humanos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrogênese/efeitos dos fármacos , Meios de Cultura/química , Meios de Cultura/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Soroalbumina Bovina/farmacologia , Soroalbumina Bovina/química , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
5.
Int Orthop ; 37(11): 2297-303, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23860792

RESUMO

PURPOSE: Our aim was to evaluate the impact of cryopreservation, cultivation time and patient's age on the expression of specific chondrogenic markers in Hyalograft® C transplants. METHODS: Gene expression of chondrocyte markers [collagen type I (COL1A1), COL2A1, aggrecan, versican, melanoma inhibitory activity (MIA) and interleukin (IL)-1ß] was analysed in cartilage biopsies (n = 17) and Hyalograft® C transplant samples (non-cryopreserved = 78, cryopreserved = 13) by quantitative real-time polymerase chain reaction (PCR). Correlation analyses were performed to evaluate the influence of the above-named parameters on the level of gene expression. RESULTS: Cryopreservation of cells was found to decrease COL2A1 and MIA significantly (4.6-fold, p < 0.01 and 2-fold, p < 0.045, respectively). The duration of cryopreservation had no further influence on the expression of these factors. No correlation was detected between cultivation time (75 ± 31 days) and the expression level of any gene. Cartilage transplants from older patients (>35 years) exhibited a significantly higher IL-1ß expression (3.7-fold, p < 0.039) than transplants from younger patients (≤ 35 years). CONCLUSIONS: Our data demonstrate that cryopreservation has a profound impact on chondrocyte metabolic activity by decreasing the expression of COL2A1 and MIA in Hyalograft® C transplants, independent of the duration of cryopreservation.


Assuntos
Transplante Ósseo/métodos , Cartilagem Articular/metabolismo , Condrócitos/transplante , Condrogênese/genética , Condrogênese/fisiologia , Criopreservação/métodos , Expressão Gênica/fisiologia , Adulto , Fatores Etários , Agrecanas/genética , Agrecanas/metabolismo , Cartilagem Articular/citologia , Condrócitos/citologia , Condrócitos/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Expressão Gênica/genética , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fatores de Tempo , Engenharia Tecidual , Versicanas/genética , Versicanas/metabolismo
6.
Methods Mol Biol ; 2598: 301-311, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36355300

RESUMO

Removing cellular material from a tissue, a process called decellularization, reduces the risk of adverse host reactions, allows for efficient decontamination, and extends the shelf-life of the matrix. It facilitates the use of cartilage tissue as human-derived allograft, thus providing the field of cartilage regeneration with a biomaterial unmatched in its similarity to native cartilage in terms of structure, composition, and mechanical properties.The dense extracellular matrix of articular cartilage requires a particularly thorough process to achieve the removal of cells, cell debris, and reagents used in the process. In our studies (Nürnberger et al., EBioMedicine 64:103196, 2021; Schneider et al., Tissue Eng Part C Methods 22(12):1095-1107, 2016), we have successfully developed a protocol for achieving decellularization via physical, chemical, and enzymatic steps. Combining freeze-thaw cycles for devitalization, hydrochloric acid as decellularization agent and the enzymatic removal of glycosaminoglycans, results in an acellular scaffold that is fully biocompatible and promotes cellular attachment. The structure and sophisticated architecture of collagen type II is left intact.This chapter provides a comprehensive guide to the steps and reagents needed to decellularize articular cartilage. In addition to the standard decell-deGAG protocol, a fast option is given which is suitable for thin specimen. Histological evaluation is presented to illustrate treatment success.


Assuntos
Cartilagem Articular , Humanos , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Ácido Clorídrico , Matriz Extracelular/química
7.
Am J Physiol Gastrointest Liver Physiol ; 303(12): G1373-83, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23064756

RESUMO

It is well known that systemic inflammatory response (SIR) often causes liver dysfunction. The aim of this study was to identify the intracellular compartment in the liver most susceptible to SIR. We analyzed morphology, ultrastructure, proteome, and expression of relevant genes in livers of rats subjected to endotoxic shock. Histological examination revealed that focal necrosis in liver was insignificant to explain liver dysfunction. Electron microscopy revealed no morphological changes in the mitochondrial structure and in the cytosol, but dilated endoplasmic reticulum (ER) cisterns were frequently observed. Apoptosis was found in white blood cells within liver tissue but not in hepatocytes. Mitochondrial, ER, and cytosolic fractions were subjected to proteome analysis by difference gel electrophoresis, and the protein spots with the highest degree of differential regulation were identified with mass spectrometry. The most pronounced proteome changes appeared in the ER, manifested as a remarkable downregulation of several proteins essential for ER functions, such as protein synthesis and transport, whereas the changes in mitochondrial and cytosolic fractions suggested a compensatory response. ER stress, as an underlying mechanism for ER impairment, was confirmed by analysis of upstream (splicing X-box-binding protein 1 mRNA) and downstream (e.g., 78-kDa glucose-regulated protein mRNA) markers, suggesting ongoing unresolved ER stress as a cause for ER dilation. Because ER is the intracellular compartment responsible for the major liver functions, our data suggest that inflammatory mediators induce unresolved ER stress, resulting in the biochemical, functional, and morphological impairment of ER that in turn causes liver dysfunction. The pathway activating ER stress in response to SIR is not known yet.


Assuntos
Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Hepatite/metabolismo , Fígado/metabolismo , Fígado/patologia , Proteoma/metabolismo , Animais , Apoptose , Masculino , Ratos , Ratos Sprague-Dawley , Síndrome de Resposta Inflamatória Sistêmica
8.
PLoS One ; 17(1): e0262294, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34986173

RESUMO

PURPOSE: The aim of this study was to investigate the effect of extracorporeal shockwave therapy (ESWT) on bone microstructure as well as the bone-tendon-interface and the musculo-tendinous transition zone to explain the previously shown improved biomechanics in a degenerative rotator cuff tear animal model. This study hypothesized that biomechanical improvements related to ESWT are a result of improved bone microstructure and muscle tendon properties. METHODS: In this controlled laboratory study unilateral supraspinatus (SSP) tendon detachment was performed in 48 male Sprague-Dawley rats. After a degeneration period of three weeks, SSP tendon was reconstructed transosseously. Rats were randomly assigned into three groups (n = 16 per group): control (noSW); intraoperative shockwave treatment (IntraSW); intra- and postoperative shockwave treatment (IntraPostSW). Eight weeks after SSP repair, all rats were sacrificed and underwent bone microstructure analysis as well as histological and immunohistochemical analyses. RESULTS: With exception of cortical porosity at the tendon area, bone microstructure analyses revealed no significant differences between the three study groups regarding cortical and trabecular bone parameters. Cortical Porosity at the Tendon Area was lowest in the IntraPostSW (p≤0.05) group. Histological analyses showed well-regenerated muscle and tendon structures in all groups. Immunohistochemistry detected augmented angiogenesis at the musculo-tendinous transition zone in both shockwave groups indicated by CD31 positive stained blood vessels. CONCLUSION: In conclusion, bone microarchitecture changes are not responsible for previously described improved biomechanical results after shockwave treatment in rotator cuff repair in rodents. Immunohistochemical analysis showed neovascularization at the musculo-tendinous transition zone within ESWT-treated animals. Further studies focusing on neovascularization at the musculo-tendinous transition zone are necessary to explain the enhanced biomechanical and functional properties observed previously. CLINICAL RELEVANCE: In patients treated with a double-row SSP tendon repair, an improvement in healing through ESWT, especially in this area, could prevent a failure of the medial row, which is considered a constantly observed tear pattern.


Assuntos
Fenômenos Biomecânicos/fisiologia , Osso Esponjoso/fisiologia , Lesões do Manguito Rotador/terapia , Manguito Rotador/fisiologia , Cicatrização/fisiologia , Animais , Artroplastia/métodos , Osso Esponjoso/cirurgia , Modelos Animais de Doenças , Tratamento por Ondas de Choque Extracorpóreas/métodos , Masculino , Ratos , Ratos Sprague-Dawley , Procedimentos de Cirurgia Plástica/métodos , Manguito Rotador/cirurgia , Lesões do Manguito Rotador/fisiopatologia , Lesões do Manguito Rotador/cirurgia , Ruptura/fisiopatologia , Ruptura/cirurgia , Ruptura/terapia , Tendões/fisiologia , Tendões/cirurgia , Microtomografia por Raio-X/métodos
9.
Cartilage ; 13(1): 19476035221075951, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35189712

RESUMO

OBJECTIVE: Implantation of tissue-engineered tracheal grafts represents a visionary strategy for the reconstruction of tracheal wall defects after resections and may develop into a last chance for a number of patients with severe cicatricial stenosis. The use of a decellularized tracheal substrate would offer an ideally stiff graft, but the matrix density would challenge efficient remodeling into a living cartilage. In this study, we hypothesized that the pores of decellularized laser-perforated tracheal cartilage (LPTC) tissues can be colonized by adult nasal chondrocytes (NCs) to produce new cartilage tissue suitable for the repair of tracheal defects. DESIGN: Human, native tracheal specimens, isolated from cadaveric donors, were exposed to decellularized and laser engraving-controlled superficial perforation (300 µm depth). Human or rabbit NCs were cultured on the LPTCs for 1 week. The resulting revitalized tissues were implanted ectopically in nude mice or orthotopically in tracheal wall defects in rabbits. Tissues were assayed histologically and by microtomography analyses before and after implantation. RESULTS: NCs were able to efficiently colonize the pores of the LPTCs. The extent of colonization (i.e., percentage of viable cells spanning >300 µm of tissue depth), cell morphology, and cartilage matrix deposition improved once the revitalized constructs were implanted ectopically in nude mice. LPTCs could be successfully grafted onto the tracheal wall of rabbits without any evidence of dislocation or tracheal stenosis, 8 weeks after implantation. Rabbit NCs, within the LPTCs, actively produced new cartilage matrix. CONCLUSION: Implantation of NC-revitalized LPTCs represents a feasible strategy for the repair of tracheal wall defects.


Assuntos
Gravuras e Gravação , Engenharia Tecidual , Animais , Cartilagem/transplante , Humanos , Lasers , Camundongos , Camundongos Nus , Coelhos , Engenharia Tecidual/métodos , Alicerces Teciduais
10.
J Tissue Eng Regen Med ; 16(2): 207-222, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34861104

RESUMO

Cartilage damage typically starts at its surface, either due to wear or trauma. Treatment of these superficial defects is important in preventing degradation and osteoarthritis. Biomaterials currently used for deep cartilage defects lack appropriate properties for this application. Therefore, we investigated photo-crosslinked gelatin methacryloyl (gelMA) as a candidate for treatment of surface defects. It allows for liquid application, filling of surface defects and forming a protective layer after UV-crosslinking, thereby keeping therapeutic cells in place. gelMA and photo-initiator lithium phenyl-2,4,6-trimethyl-benzoylphosphinate (Li-TPO) concentration were optimized for application as a carrier to create a favorable environment for human articular chondrocytes (hAC). Primary hAC were used in passages 3 and 5, encapsulated into two different gelMA concentrations (7.5 wt% (soft) and 10 wt% (stiff)) and cultivated for 3 weeks with TGF-ß3 (0, 1 and 10 ng/mL). Higher TGF-ß3 concentrations induced spherical cell morphology independent of gelMA stiffness, while low TGF-ß3 concentrations only induced rounded morphology in stiff gelMA. Gene expression did not vary across gel stiffnesses. As a functional model gelMA was loaded with two different cell types (hAC and/or human adipose-derived stem cells [ASC/TERT1]) and applied to human osteochondral osteoarthritic plugs. GelMA attached to the cartilage, smoothened the surface and retained cells in place. Resistance against shear forces was tested using a tribometer, simulating normal human gait and revealing maintained cell viability. In conclusion gelMA is a versatile, biocompatible material with good bonding capabilities to cartilage matrix, allowing sealing and smoothening of superficial cartilage defects while simultaneously delivering therapeutic cells for tissue regeneration.


Assuntos
Condrócitos , Engenharia Tecidual , Cartilagem/metabolismo , Gelatina/metabolismo , Gelatina/farmacologia , Humanos , Hidrogéis/farmacologia , Metacrilatos
11.
Nanotechnology ; 22(14): 145101, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21346306

RESUMO

Magnetic iron oxide nanoparticles (Fe-NP) are currently considered for various diagnostic and therapeutic applications in the brain. However, little is known on the accumulation and biocompatibility of such particles in brain cells. We have synthesized and characterized dimercaptosuccinic acid (DMSA) coated Fe-NP and have investigated their uptake by cultured brain astrocytes. DMSA-coated Fe-NP that were dispersed in physiological medium had an average hydrodynamic diameter of about 60 nm. Incubation of cultured astrocytes with these Fe-NP caused a time- and concentration-dependent accumulation of cellular iron, but did not lead within 6 h to any cell toxicity. After 4 h of incubation with 100-4000 µM iron supplied as Fe-NP, the cellular iron content reached levels between 200 and 2000 nmol mg⁻¹ protein. The cellular iron content after exposure of astrocytes to Fe-NP at 4 °C was drastically lowered compared to cells that had been incubated at 37 °C. Electron microscopy revealed the presence of Fe-NP-containing vesicles in cells that were incubated with Fe-NP at 37 °C, but not in cells exposed to the nanoparticles at 4 °C. These data demonstrate that cultured astrocytes efficiently take up DMSA-coated Fe-NP in a process that appears to be saturable and strongly depends on the incubation temperature.


Assuntos
Astrócitos/metabolismo , Encéfalo/citologia , Endocitose , Nanopartículas de Magnetita , Succímero/química , Succímero/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Sobrevivência Celular , Células Cultivadas , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestrutura , Espaço Intracelular/metabolismo , Ferro/metabolismo , Cinética , Luz , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestrutura , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Ratos , Ratos Wistar , Espalhamento de Radiação , Espectrometria por Raios X , Eletricidade Estática , Temperatura
12.
Biofabrication ; 13(3)2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33513590

RESUMO

Scaffold-free 3D cell cultures (e.g. pellet cultures) are widely used in medical science, including cartilage regeneration. Their drawbacks are high time/reagent consumption and lack of early readout parameters. While optimisation was achieved by automation or simplified spheroid generation, most culture systems remain expensive or require tedious procedures. The aim of this study was to establish a system for resource efficient spheroid generation with additional early readout parameters. This was achieved by a new approach for spheroid generation via self-assembly from monolayer via compartmentation of cell culture surfaces utilising laser engraving (grid plates). The compartmentation triggered contraction and rolling up of the cell monolayer, finishing in condensation into a spheroid in human adipose-derived stem cell (ASC/TERT1) and human articular chondrocytes (hACs)-ASC/TERT1 co-cultures, when cultivated on grid plates under chondrogenic conditions. Plates with 1 and 3 mm grid size yielded stable diameters (about 140µm and 300µm, respectively). ASC/TERT1 spheroids fully formed within 3 weeks while co-cultures took 1-2 weeks, forming significantly faster with increasing hAC ratio (p< 0.05 and 0.01 for 1:1 and 1:4 ASC/TERT1:hAC ratio, respectively). Co-cultures showed slightly lower spheroid diameters, due to earlier spheroid formation and incomplete monolayer formation. However, this was associated with a more homogeneous matrix distribution in the co-culture. Both showed differentiation capacity comparable to standard pellet culture in (immune-)histochemistry and RT-qPCR. To assess usability for cartilage repair, spheroids were embedded into a hydrogel (fibrin), yielding cellular outgrowth and matrix deposition, which was especially pronounced in co-cultures. The herein presented novel cell culture system is not only a promising tool for autonomous spheroid generation with the potential of experimental and clinical application in tissue engineering, but also for the generation of 'building blocks' for subsequential biofabrication strategies such as bioprinting.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Condrogênese , Diferenciação Celular , Células Cultivadas , Condrócitos , Humanos , Engenharia Tecidual
13.
Biochim Biophys Acta ; 1792(6): 521-30, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19327397

RESUMO

Inflammatory response has recently been shown to induce endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), which either recovers proper ER function or activates apoptosis. Here we show that endotoxin (lipopolysaccharide = LPS) can lead to functional ER failure tentatively via a mitochondrion-dependent pathway in livers of rats. Histological examination did not reveal significant damage to liver in form of necroses. Electron microscopy displayed transparent rings appearing around morphologically unchanged mitochondria, which were identified as dilated ER. The spliced mRNA variant of X-box protein-1 (XBP1) and also the mRNA of 78 kDa glucose-regulated protein (GRP78) were up-regulated, both typical markers of ER stress. However, GRP78 was down-regulated at the protein level. A pro-apoptotic shift in the bax/bcl-XL mRNA ratio was not accompanied by translocation of apoptosis inducing factor (AIF) to the nucleus, suggesting that the cells entered a pre-apoptotic state, but apoptosis was not executed. Monooxygenase activity of p450, representing the detoxification system in ER, was decreased after administration of endotoxin. Biochemical analysis of proteins important for ER function revealed the impairment of protein folding, transport, and detoxification suggesting functional ER failure. We suggest that functional ER failure may be a reason for organ dysfunction upon excessive inflammatory response mediated by endotoxin.


Assuntos
Retículo Endoplasmático/fisiologia , Inflamação/fisiopatologia , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Mitocôndrias/fisiologia , Dobramento de Proteína , Animais , Expressão Gênica , Inflamação/patologia , Fígado/patologia , Masculino , Modelos Biológicos , Estresse Oxidativo , Transporte Proteico , Ratos
14.
Artigo em Inglês | MEDLINE | ID: mdl-32426347

RESUMO

Biomechanical cues such as shear stress, stretching, compression, and matrix elasticity are vital in the establishment of next generation physiological in vitro tissue models. Matrix elasticity, for instance, is known to guide stem cell differentiation, influence healing processes and modulate extracellular matrix (ECM) deposition needed for tissue development and maintenance. To better understand the biomechanical effect of matrix elasticity on the formation of articular cartilage analogs in vitro, this study aims at assessing the redifferentiation capacity of primary human chondrocytes in three different hydrogel matrices of predefined matrix elasticities. The hydrogel elasticities were chosen to represent a broad spectrum of tissue stiffness ranging from very soft tissues with a Young's modulus of 1 kPa up to elasticities of 30 kPa, representative of the perichondral-space. In addition, the interplay of matrix elasticity and transforming growth factor beta-3 (TGF-ß3) on the redifferentiation of primary human articular chondrocytes was studied by analyzing both qualitative (viability, morphology, histology) and quantitative (RT-qPCR, sGAG, DNA) parameters, crucial to the chondrotypic phenotype. Results show that fibrin hydrogels of 30 kPa Young's modulus best guide chondrocyte redifferentiation resulting in a native-like morphology as well as induces the synthesis of physiologic ECM constituents such as glycosaminoglycans (sGAG) and collagen type II. This comprehensive study sheds light onto the mechanobiological impact of matrix elasticity on formation and maintenance of articular cartilage and thus represents a major step toward meeting the need for advanced in vitro tissue models to study both re- and degeneration of articular cartilage.

15.
J Orthop Res ; 38(11): 2464-2473, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32167192

RESUMO

Infections related to orthopedic procedures are considered particularly severe when implantation materials are used, because effective treatments for biofilm removal are lacking. In this study, the relatively new approach for infection control by using an erbium:yttrium-aluminum-garnet (Er:YAG) laser was tested. This laser vaporizes all water containing cells in a very effective, precise, and predictable manner and results in only minimal thermal damage. For preliminary testing, 42 steel plates and 42 pins were seeded with mixed cultures. First, the minimally necessary laser energy for biofilm removal was determined. Subsequently, the effectiveness of biofilm removal with the Er:YAG laser and the cleansing of the metal implants with octenidine-soaked gauze was compared. Then, we compared the effectiveness of biofilm removal on 207 steel pins from 41 patients directly after explantation. Sonication and scanning electron microscopy were used for analysis. Laser fluences exceeding 2.8 J/cm2 caused a complete extinction of all living cells by a single-laser impulse. Cleansing with octenidine-soaked gauze and irradiation with the Er:YAG laser are both thoroughly effective when applied to seeded pins. In contrast, when explanted pins with fully developed biofilms were analyzed, we found a significant advantage of the laser procedure. The Er:YAG laser offers a secure, complete, and nontoxic eradication of all kinds of pathogens from metal implants without damaging the implant and without the possible development of resistance. The precise noncontact removal of adjacent tissue is a decisive advantage over conventional disinfectants. Therefore, laser irradiation could become a valuable method in every debridement, antibiotics, and implant retention procedure.


Assuntos
Desinfecção/métodos , Lasers de Estado Sólido/uso terapêutico , Infecções Relacionadas à Prótese/terapia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biofilmes/efeitos da radiação , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
Contrast Media Mol Imaging ; 2019: 7483745, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31049044

RESUMO

Microcomputed tomography (µCT) is widely used for the study of mineralized tissues, but a similar use for soft tissues is hindered by their low X-ray attenuation. This limitation can be overcome by the recent development of different staining techniques. Staining with Lugol's solution, a mixture of one part iodine and two parts potassium iodide in water, stands out among these techniques for its low complexity and cost. Currently, Lugol staining is mostly used for anatomical examination of tissues. In the present study, we seek to optimize the quality and reproducibility of the staining for ex vivo visualization of soft tissues in the context of a peripheral nerve regeneration model in the rat. We show that the staining result not only depends on the concentration of the staining solution but also on the amount of stain in relation to the tissue volume and composition, necessitating careful adaptation of the staining protocol to the respective specimen tissue. This optimization can be simplified by a stepwise staining which we show to yield a similar result compared to staining in a single step. Lugol staining solution results in concentration-dependent tissue shrinkage which can be minimized but not eliminated. We compared the shrinkage of tendon, nerve, skeletal muscle, heart, brain, and kidney with six iterations of Lugol staining. 60 ml of 0.3% Lugol's solution per cm3 of tissue for 24 h yielded good results on the example of a peripheral nerve regeneration model, and we were able to show that the regenerating nerve inside a silk fibroin tube can be visualized in 3D using this staining technique. This information helps in deciding the region of interest for histological imaging and provides a 3D context to histological findings. Correlating both imaging modalities has the potential to improve the understanding of the regenerative process.


Assuntos
Iodo/farmacologia , Sistema Musculoesquelético/diagnóstico por imagem , Regeneração Nervosa/fisiologia , Nervos Periféricos/diagnóstico por imagem , Animais , Meios de Contraste/farmacologia , Humanos , Imageamento Tridimensional/métodos , Sistema Musculoesquelético/patologia , Nervos Periféricos/crescimento & desenvolvimento , Nervos Periféricos/patologia , Ratos , Microtomografia por Raio-X/métodos
17.
Int J Mol Med ; 22(5): 691-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18949392

RESUMO

Matrix-associated autologous chondrocyte transplantation (MACT) is a tissue-engineered approach for the treatment of cartilage defects and combines autologous chondrocytes seeded on biomaterials. The objective of the study is the analysis of growth and differentiation behaviour of human articular chondrocytes grown on three different matrices used for MACT. Human articular chondrocytes were kept in monolayer culture for 42 days and then seeded on matrices consisting of either collagen type I/III, hyaluronan, or gelatine. During the culture time of 4 weeks the constructs were analyzed weekly. Morphological criteria were studied by scanning and transmission electron microscopy. The expression of the main type collagens was analyzed by real-time PCR. The collagen type I/III matrix supported a differentiation that closely resembled the tissue organisation of native cartilage, but cell number and type II collagen synthesis were low and differentiation occurred rather late in the cultivation period. The hyaluronan matrix and the gelatine-based matrix supported a rather rapid differentiation, with a high number of cells and a relatively high amount of type II collagen, but there was no spatial assembly that mimicked native cartilage. These facts indicate that the nature of the matrix is of great influence in the differentiation behaviour of dedifferentiated chondrocytes.


Assuntos
Materiais Biocompatíveis , Cartilagem Articular/metabolismo , Diferenciação Celular , Condrócitos/metabolismo , Doenças das Cartilagens/patologia , Doenças das Cartilagens/terapia , Cartilagem Articular/ultraestrutura , Células Cultivadas , Condrócitos/transplante , Condrócitos/ultraestrutura , Colágeno Tipo I/química , Colágeno Tipo II/biossíntese , Colágeno Tipo III/química , Gelatina/química , Humanos , Ácido Hialurônico/química , Fatores de Tempo
18.
Biol Rev Camb Philos Soc ; 93(2): 1056-1076, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29119723

RESUMO

The majority of ticks in the family Ixodidae secrete a substance anchoring their mouthparts to the host skin. This substance is termed cement. It has adhesive properties and seals the lesion during feeding. The particular chemical composition and the curing process of the cement are unclear. This review summarizes the literature, starting with a historical overview, briefly introducing the different hypotheses on the origin of the adhesive and how the tick salivary glands have been identified as its source. Details on the sequence of cement deposition, the curing process and detachment are provided. Other possible functions of the cement, such as protection from the host immune system and antimicrobial properties, are presented. Histochemical and ultrastructural data of the intracellular granules in the salivary gland cells, as well as the secreted cement, suggest that proteins constitute the main material, with biochemical data revealing glycine to be the dominant amino acid. Applied methods and their restrictions are discussed. Tick cement is compared with adhesives of other animals such as barnacles, mussels and sea urchins. Finally, we address the potential of tick cement for the field of biomaterial research and in particular for medical applications in future.


Assuntos
Proteínas de Artrópodes/química , Proteínas de Artrópodes/fisiologia , Produtos Biológicos , Carrapatos/fisiologia , Animais , Comportamento Alimentar , Humanos , Glândulas Salivares/fisiologia
19.
Artigo em Inglês | MEDLINE | ID: mdl-29468155

RESUMO

There is continual demand for animal models that allow a quantitative assessment of angiogenic properties of biomaterials, therapies, and pharmaceuticals. In its simplest form, this is done by subcutaneous material implantation and subsequent vessel counting which usually omits spatial data. We have refined an implantation model and paired it with a computational analytic routine which outputs not only vessel count but also vessel density, distribution, and vessel penetration depth, that relies on a centric vessel as a reference point. We have successfully validated our model by characterizing the angiogenic potential of a fibrin matrix in conjunction with recombinant human vascular endothelial growth factor (rhVEGF165). The inferior epigastric vascular pedicles of rats were sheathed with silicone tubes, which were subsequently filled with 0.2 ml of fibrin and different doses of rhVEGF165, centrically embedding the vessels. Over 4 weeks, tissue samples were harvested and subsequently immunohistologically stained and computationally analyzed. The model was able to detect variations over the angiogenic potentials of growth factor spiked fibrin matrices. Adding 20 ng of rhVEGF165 resulted in a significant increase in vasculature while 200 ng of rhVEGF165 did not improve vascular growth. Vascularized tissue volume increased during the first week and vascular density increased during the second week. Total vessel count increased significantly and exhibited a peak after 2 weeks which was followed by a resorption of vasculature by week 4. In summary, a simple implantation model to study in vivo vascularization with only a minimal workload attached was enhanced to include morphologic data of the emerging vascular tree.

20.
Sci Rep ; 8(1): 17010, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30451865

RESUMO

Osteoarthritis (OA) is one of the most common causes of disability and represents a major socio-economic burden. Despite intensive research, the molecular mechanisms responsible for the initiation and progression of OA remain inconclusive. In recent years experimental findings revealed elevated levels of reactive oxygen species (ROS) as a major factor contributing to the onset and progression of OA. Hence, we designed a hydrostatic pressure bioreactor system that is capable of stimulating cartilage cell cultures with elevated ROS levels. Increased ROS levels in the media did not only lead to an inhibition of glycosaminoglycans and collagen II formation but also to a reduction of already formed glycosaminoglycans and collagen II in chondrogenic mesenchymal stem cell pellet cultures. These effects were associated with the elevated activity of matrix metalloproteinases as well as the increased expression of several inflammatory cytokines. ROS activated different signaling pathways including PI3K/Akt and MAPK/ERK which are known to be involved in OA initiation and progression. Utilizing the presented bioreactor system, an OA in vitro model based on the generation of ROS was developed that enables the further investigation of ROS effects on cartilage degradation but can also be used as a versatile tool for anti-oxidative drug testing.


Assuntos
Cartilagem Articular/patologia , Condrogênese , Pressão Hidrostática/efeitos adversos , Células-Tronco Mesenquimais/patologia , Osteoartrite/etiologia , Espécies Reativas de Oxigênio/metabolismo , Cartilagem Articular/metabolismo , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/metabolismo , Osteoartrite/patologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA