Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomater Sci ; 11(20): 6801-6822, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37622217

ABSTRACT

An important challenge in tissue engineering is the regeneration of functional articular cartilage (AC). In the field, biomimetic hydrogels are being extensively studied as scaffolds that recapitulate microenvironmental features or as mechanical supports for transplanted cells. New advanced hydrogel formulations based on salmon methacrylate gelatin (sGelMA), a cold-adapted biomaterial, are presented in this work. The psychrophilic nature of this biomaterial provides rheological advantages allowing the fabrication of scaffolds with high concentrations of the biopolymer and high mechanical strength, suitable for formulating injectable hydrogels with high mechanical strength for cartilage regeneration. However, highly intricate cell-laden scaffolds derived from highly concentrated sGelMA solutions could be deleterious for cells and scaffold remodeling. On this account, the current study proposes the use of sGelMA supplemented with a mesophilic sacrificial porogenic component. The cytocompatibility of different sGelMA-based formulations is tested through the encapsulation of osteoarthritic chondrocytes (OACs) and stimulated to synthesize extracellular matrix (ECM) components in vitro and in vivo. The sGelMA-derived scaffolds reach high levels of stiffness, and the inclusion of porogens impacts positively the scaffold degradability and molecular diffusion, improved fitness of OACs, increased the expression of cartilage-related genes, increased glycosaminoglycan (GAG) synthesis, and improved remodeling toward cartilage-like tissues. Altogether, these data support the use of sGelMA solutions in combination with mammalian solid gelatin beads for highly injectable formulations for cartilage regeneration, strengthening the importance of the balance between mechanical properties and remodeling capabilities.


Subject(s)
Cartilage, Articular , Gelatin , Animals , Porosity , Chondrocytes/transplantation , Tissue Engineering , Hydrogels , Biocompatible Materials , Regeneration , Tissue Scaffolds , Mammals
2.
Biomed Mater ; 18(4)2023 05 24.
Article in English | MEDLINE | ID: mdl-37167997

ABSTRACT

Although there have been many advances in injectable hydrogels as scaffolds for tissue engineering or as payload-containing vehicles, the lack of adequate microporosity for the desired cell behavior, tissue integration, and successful tissue generation remains an important drawback. Herein, we describe an effective porous injectable system that allowsin vivoformation of pores through conventional syringe injection at room temperature. This system is based on the differential melting profiles of photocrosslinkable salmon gelatin and physically crosslinked porogens of porcine gelatin (PG), in which PG porogens are solid beads, while salmon methacrylamide gelatin remains liquid during the injection procedure. After injection and photocrosslinking, the porogens were degraded in response to the physiological temperature, enabling the generation of a homogeneous porous structure within the hydrogel. The resultant porogen-containing formulations exhibited controlled gelation kinetics within a broad temperature window (18.5 ± 0.5-28.8 ± 0.8 °C), low viscosity (133 ± 1.4-188 ± 16 cP), low force requirements for injectability (17 ± 0.3-39 ± 1 N), robust mechanical properties after photo-crosslinking (100.9 ± 3.4-332 ± 13.2 kPa), and favorable cytocompatibility (>70% cell viability). Remarkably,in vivosubcutaneous injection demonstrated the suitability of the system with appropriate viscosity and swift crosslinking to generate porous hydrogels. The resulting injected porous constructs showed favorable biocompatibility and facilitated cell infiltration for desirable potential tissue remodeling. Finally, the porogen-containing formulations exhibited favorable handling, easy deposition, and good shape fidelity when used as bioinks in 3D bioprinting technology. This injectable porous system serves as a platform for various biomedical applications, thereby inspiring future advances in cell therapy and tissue engineering.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Gelatin/chemistry , Porosity , Biocompatible Materials/chemistry , Hydrogels/chemistry , Printing, Three-Dimensional
3.
Biomacromolecules ; 24(1): 150-165, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36542545

ABSTRACT

The increasing demand for tissue replacement has encouraged scientists worldwide to focus on developing new biofabrication technologies. Multimaterials/cells printed with stringent resolutions are necessary to address the high complexity of tissues. Advanced inkjet 3D printing can use multimaterials and attain high resolution and complexity of printed structures. However, a decisive yet limiting aspect of translational 3D bioprinting is selecting the befitting material to be used as bioink; there is a complete lack of cytoactive bioinks with adequate rheological, mechanical, and reactive properties. This work strives to achieve the right balance between resolution and cell support through methacrylamide functionalization of a psychrophilic gelatin and new fluorosurfactants used to engineer a photo-cross-linkable and immunoevasive bioink. The syntonized parameters following optimal formulation conditions allow proficient printability in a PolyJet 3D printer comparable in resolution to a commercial synthetic ink (∼150 µm). The bioink formulation achieved the desired viability (∼80%) and proliferation of co-printed cells while demonstrating in vivo immune tolerance of printed structures. The practical usage of existing high-resolution 3D printing systems using a novel bioink is shown here, allowing 3D bioprinted structures with potentially unprecedented complexity.


Subject(s)
Bioprinting , Bioprinting/methods , Printing, Three-Dimensional , Gelatin/chemistry , Rheology , Tissue Scaffolds/chemistry , Tissue Engineering/methods
4.
Front Psychol ; 12: 652884, 2021.
Article in English | MEDLINE | ID: mdl-33935914

ABSTRACT

Positive family functioning during adolescence is usually studied analyzing parents' competences and children's relationship abilities. We present an instrument for the assessment of parental competence, which encompasses both educational style and transmission of values. The objective of the study was to analyze its factor structure and to assess its value in predicting adolescent outcomes. We recruited 2,459 high school students, aged 12-15, in 40 schools from Spain, Peru, Mexico, and Chile. They responded to the instrument and to other questions regarding lifestyles. Exploratory and confirmatory factor analyses were carried out in order to assess the internal structure of the instrument, and internal consistency of the resulting dimensions was calculated. Finally, regression analyses were performed to establish associations between the parenting dimensions and adolescent outcomes. Factor analyses showed a consistent structure, with good fit indices in the four countries. The final four factors include parenting styles (Warmth and Demandingness) and education in values (Fortitude and Privacy). Regression analyses showed associations with adolescent outcomes. For example, adolescents' life satisfaction was best predicted by Warmth and Education in fortitude. Practical implications are suggested.

5.
Front Pharmacol ; 11: 679, 2020.
Article in English | MEDLINE | ID: mdl-32528280

ABSTRACT

The clinical benefit of therapies using Mesenchymal Stem Cells (MSCs) is attributable to their pleiotropic effect over cells and tissues, mainly through their secretome. This paracrine effect is mediated by secreted growth factors and extracellular vesicles (EV) including small EV (sEV). sEV are extra-cellular, membrane encompassed vesicles of 40 to 200 nm diameter that can trigger and signal many cellular responses depending on their cargo protein and nucleic acid repertoire. sEV are purified from cell culture conditioned media using several kits and protocols available that can be tedious and time-consuming, involving sequences of ultracentrifugations and density gradient separations, making their production a major challenge under Good Manufacturing Practices (GMP) conditions. We have developed a method to efficiently enrich cell culture media with high concentrations of sEV by encapsulating cells in semipermeable cellulose beads that allows selectively the release of small particles while offering a 3D culture condition. This method is based on the pore size of the capsules, allowing the release of particles of ≤ 200 nm including sEV. As a proof-of-principle, MSCs were encapsulated and their sEV release rate (sEV-Cap) was monitored throughout the culture and compared to sEV isolated from 2D seeded cells (sEV-2D) by repetitive ultracentrifugation cycles or a commercial kit. The isolated sEV expressed CD63, CD9, and CD81 as confirmed by flow cytometry analysis. Under transmission electron microscopy (TEM), they displayed the similar rounded morphology as sEV-2D. Their corresponding diameter size was validated by nanoparticle tracking analysis (NTA). Interestingly, sEV-Cap retained the expected biological activities of MSCs, including a pro-angiogenic effect over endothelial cells, neuritic outgrowth stimulation in hippocampal neurons and immunosuppression of T cells in vitro. Here, we successfully present a novel, cost, and time-saving method to generate sEV from encapsulated MSCs. Future applications include using encapsulated cells as a retrievable delivery device that can interact with the host niche by releasing active agents in vivo, including sEV, growth factors, hormones, and small molecules, while avoiding cell clearance, and the negative side-effect of releasing undesired components including apoptotic bodies. Finally, particles produced following the encapsulation protocol display beneficial features for their use as drug-loaded delivery vehicles.

6.
J Exp Biol ; 222(Pt 20)2019 10 16.
Article in English | MEDLINE | ID: mdl-31537653

ABSTRACT

Microvesicles are key players in cellular communication. As glandular secretions present a rich source of active exosomes, we hypothesized that exosome-like vesicles are present in Apis mellifera hypopharyngeal gland secretomal products (honey, royal jelly and bee pollen), and participate in their known antibacterial and pro-regenerative effects. We developed an isolation protocol based on serial centrifugation and ultracentrifugation steps and demonstrated the presence of protein-containing exosome-like vesicles in all three bee-derived products. Assessing their antibacterial properties, we found that exosome-like vesicles had bacteriostatic, bactericidal and biofilm-inhibiting effects on Staphylococcus aureus Furthermore, we demonstrated that mesenchymal stem cells (MSCs) internalize bee-derived exosome-like vesicles and that these vesicles influence the migration potential of the MSCs. In an in vitro wound-healing assay, honey and royal jelly exosome-like vesicles increased migration of human MSCs, demonstrating their inter-kingdom activity. In summary, we have discovered exosome-like vesicles as a new, active compound in bee pollen, honey and royal jelly.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bees/metabolism , Exosomes/metabolism , Fatty Acids/chemistry , Honey , Pollen/chemistry , Regeneration/drug effects , Animals , Cell Movement/drug effects , Endocytosis/drug effects , Exosomes/drug effects , Humans , Pollen/ultrastructure
7.
Stem Cell Res Ther ; 10(1): 249, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399041

ABSTRACT

BACKGROUND: The generation of functional human epidermal melanocytes (HEM) from stem cells provides an unprecedented source for cell-based therapy in vitiligo. Despite the important efforts exerted to obtain melanin-producing cells from stem cells, pre-clinical results still lack the safety and scalability characteristics essential for their translational application. METHODS: Here, we report a rapid and efficient protocol based on defined culture conditions capable of differentiating adult adipose-derived stem cells (ADSC) to scalable amounts of proliferative melanocyte precursors (PreMel) within 30 days. PreMel were characterized in vitro through qPCR, Western blot, flow cytometry, biochemical assays, and in vivo assays in immunocompromised mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ, or NSG). RESULTS: After 30 days of differentiation, the stem cell-derived PreMel were defined as CD105neg CD73low according to immunophenotypic changes in comparison with parental stem cell markers. In addition, expression of microphthalmia-associated transcription factor (MITF), active tyrosinase (TYR), and the terminal differentiation-involved premelanosome protein (PMEL) were detected. Furthermore, PreMel had the potential to synthesize melanin and package it into melanosomes both in vitro and in vivo in NSG mice skin. CONCLUSIONS: This study proposes a rapid and scalable protocol for the generation of proliferative melanocyte precursors (PreMel) from ADSC. These PreMel display the essential functional characteristics of bona fide HEM, opening a new path for an autologous cellular therapy for vitiligo patients.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Melanocytes/metabolism , 5'-Nucleotidase/metabolism , Adolescent , Adult , Animals , Cell Lineage , Endoglin/metabolism , Female , Humans , Melanins/metabolism , Melanocytes/cytology , Melanocytes/transplantation , Mice , Mice, Inbred NOD , Microphthalmia-Associated Transcription Factor/metabolism , Middle Aged , Monophenol Monooxygenase/metabolism , Skin/pathology , Stem Cells/cytology , Stem Cells/metabolism , Vitiligo/pathology , Vitiligo/therapy , Young Adult , gp100 Melanoma Antigen/metabolism
9.
Nat Commun ; 10(1): 3098, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31308369

ABSTRACT

Design strategies for small diameter vascular grafts are converging toward native-inspired tissue engineered grafts. A new automated technology is presented that combines a dip-spinning methodology for depositioning concentric cell-laden hydrogel layers, with an adapted solution blow spinning (SBS) device for intercalated placement of aligned reinforcement nanofibres. This additive manufacture approach allows the assembly of bio-inspired structural configurations of concentric cell patterns with fibres at specific angles and wavy arrangements. The middle and outer layers were tuned to structurally mimic the media and adventitia layers of native arteries, enabling the fabrication of small bore grafts that exhibit the J-shape mechanical response and compliance of human coronary arteries. This scalable automated system can fabricate cellularized multilayer grafts within 30 min. Grafts were evaluated by hemocompatibility studies and a preliminary in vivo carotid rabbit model. The dip-spinning-SBS technology generates constructs with native mechanical properties and cell-derived biological activities, critical for clinical bypass applications.


Subject(s)
Bioprosthesis , Blood Vessel Prosthesis , Coronary Vessels/anatomy & histology , Tissue Engineering/methods , Animals , Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis Implantation/methods , Coronary Artery Bypass/instrumentation , Coronary Artery Bypass/methods , Female , Human Umbilical Vein Endothelial Cells , Humans , Hydrogels/chemistry , Materials Testing/methods , Models, Animal , Rabbits , Tensile Strength
10.
Stem Cells Int ; 2019: 1916542, 2019.
Article in English | MEDLINE | ID: mdl-30809262

ABSTRACT

Preeclampsia is a pregnancy-specific disorder defined by the new onset of hypertension and proteinuria after 20 weeks of gestation. Although its precise etiology is still unknown, there is evidence suggesting that it may be a consequence of impaired decidual and stromal cell function. Recently, a stem cell population derived from endometrial tissue and isolated from menstrual effluent called menstrual stem cells (MenSCs) has been identified. MenSCs exhibit important angiogenic and inflammatory properties that may contribute to both normal and pathological complications of implantation and placentation, including preeclampsia. We hypothesized that the angiogenic and inflammatory activity of MenSCs is altered in women who have a past history of preeclampsia and that this phenotype persists postpartum. The primary outcome measures were stromal progenitor cell formation, in vitro induction of endothelial tube formation, and release of proinflammatory cytokines. MenSCs obtained from women with a previous normal or preeclamptic pregnancy displayed similar phenotypic characteristics, tri-differentiation capacity, and proliferation. MenSCs derived from women who had preeclampsia on their previous pregnancy had reduced angiogenic capacity (~30% fewer junctions and nodes, p < 0.05) and stromal progenitor cell formation (<50% measured at a serial dilution of 1 : 10.000, p < 0.05) when compared to controls. In vitro, MenSCs obtained from patients with a history of preeclampsia expressed less endoglin and secreted less VEGF but more IL-6 than controls did. These data are consistent with the hypothesis that the angiogenic and inflammatory properties of MenSCs of women with a previous pregnancy complicated by preeclampsia have reduced angiogenic capacity and are more proinflammatory than those of MenSCs of women with a previous normal pregnancy. This altered phenotype of MenSCs observed following preeclampsia could either be present before the development of the pathology, predisposing the endometrial milieu to and consequently leading to limited vascular remodeling, or be a consequence of preeclampsia itself. The former may afford opportunity for targeted therapeutic intervention; the latter, a putative biomarker for future risk of pregnancy complications.

11.
Stem Cell Res Ther ; 8(1): 203, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28962669

ABSTRACT

BACKGROUND: Wharton's jelly-derived mesenchymal stem cells (WJ-MSC) show remarkable therapeutic potential to repair tissue upon injury via paracrine signaling by secreting diverse trophic factors that promote angiogenesis. However, the mechanisms and signaling pathways that regulate the induction of these specific factors are still mostly unknown. Emerging evidence suggests that Sonic hedgehog (SHH) plays a central role in angiogenesis and tissue maintenance. However, its contribution to the angiogenic potential of MSC has not been fully addressed. The aim of this work was to characterize the expression of the SHH pathway components in WJ-MSC primary cultures and to evaluate their angiogenic responsiveness to SHH signaling. METHODS: Primary cell cultures obtained from human umbilical cords were treated with pharmacological modulators of the SHH pathway. We evaluated the modulation of diverse trophic factors in cell lysates, conditioned medium, and functional in vitro assays. In addition, we determined the angiogenic potential of the SHH pathway in the chicken chorioallantoic membrane, an in vivo model. RESULTS: Our results show that WJ-MSC express components of the canonical SHH pathway and are activated by its signaling. In fact, we provide evidence of basal autocrine/paracrine SHH signaling in WJ-MSC. SHH pathway stimulation promotes the secretion of angiogenic factors such as activin A, angiogenin, angiopoietin 1, granulocyte-macrophage colony-stimulating factor, matrix metallometallopeptidase -9, and urokinase-type plasminogen activator, enhancing the pro-angiogenic capabilities of WJ-MSC both in vitro and in vivo. CONCLUSION: WJ-MSC are a cell population responsive to SHH pathway stimulation. Basal SHH signaling is in part responsible for the angiogenic inductive properties of WJ-MSC. Overall, exogenous activation of the SHH pathway enhances the angiogenic properties of WJ-MSC, making this cell population an ideal target for treating tissue injury.


Subject(s)
Hedgehog Proteins/metabolism , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Activins/genetics , Activins/metabolism , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Chickens , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hedgehog Proteins/genetics , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Plasminogen Activators/genetics , Plasminogen Activators/metabolism , Signal Transduction , Wharton Jelly/cytology
12.
Angiogenesis ; 17(4): 851-66, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24728929

ABSTRACT

Disorders in skin wound healing are a major health problem that requires the development of innovative treatments. The use of biomaterials as an alternative of skin replacement has become relevant, but its use is still limited due to poor vascularization inside the scaffolds, resulting in insufficient oxygen and growth factors at the wound site. In this study, we have developed a cell-based wound therapy consisting of the application of collagen-based dermal scaffolds containing mesenchymal stem cells from Wharton's jelly (WJ-MSC) in an immunocompetent mouse model of angiogenesis. From our comparative study on the secretion profile between WJ-MSC and adipose tissue-derived MSC, we found a stronger expression of several well-characterized growth factors, such as VEGF-A, angiopoietin-1 and aFGF, which are directly linked to angiogenesis, in the culture supernatant of WJ-MSC, both on monolayer and 3D culture conditions. WJ-MSC proved to be angiogenic both in vitro and in vivo, through tubule formation and CAM assays, respectively. Moreover, WJ-MSC consistently improved the healing response in vivo in a mouse model of human-like dermal repair, by triggering angiogenesis and further providing a suitable matrix for wound repair, without altering the inflammatory response in the animals. Since these cells can be easily isolated, cultured with high expansion rates and cryopreserved, they represent an attractive stem cell source for their use in allogeneic cell transplant and tissue engineering.


Subject(s)
Mesenchymal Stem Cells/cytology , Neovascularization, Pathologic , Regeneration/physiology , Skin/metabolism , Wharton Jelly/chemistry , Adipocytes/cytology , Animals , Biocompatible Materials , Cell Proliferation , Chickens , Chorioallantoic Membrane , Cryopreservation , Culture Media, Conditioned , Flow Cytometry , Humans , Inflammation , Male , Mice , Mice, Inbred BALB C , Osteogenesis , Proteome , Skin/pathology , Tissue Engineering , Umbilical Cord/pathology , Wound Healing
13.
Biol Res ; 39(1): 189-90, 2006.
Article in English | MEDLINE | ID: mdl-16629179

ABSTRACT

Recent evidence suggests that reactive oxygen species function as second messenger molecules in normal physiological processes. For example, activation of N-Methyl-D-Aspartate receptor results in the production of ROS, which appears to be critical for synaptic plasticity, one of the cellular mechanisms that underlie learning and memory. In this work, we studied the effect of iron in the activation of MAPK/ERK pathway and on Ca2+ signaling in neuronal PC12 cells. We found that iron-dependent generation of hydroxyl radicals is likely to modulate Ca2+ signaling through RyR calcium channel activation, which, in turn, activates the MAPK/ERK pathway. These findings underline the relevance of iron in normal neuronal function.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Iron/pharmacology , Neuroblastoma/enzymology , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Animals , Blotting, Western , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , PC12 Cells/metabolism , Phosphorylation/drug effects , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects
14.
Biol. Res ; 39(1): 189-190, 2006. ilus
Article in English | LILACS | ID: lil-430712

ABSTRACT

Recent evidence suggests that reactive oxygen species function as second messenger molecules in normal physiological processes. For example, activation of N-Methyl-D-Aspartate receptor results in the production of ROS, which appears to be critical for synaptic plasticity, one of the cellular mechanisms that underlie learning and memory. In this work, we studied the effect of iron in the activation of MAPK/ERK pathway and on Ca2+ signaling in neuronal PC12 cells. We found that iron-dependent generation of hydroxyl radicals is likely to modulate Ca2+ signaling through RyR calcium channel activation, which, in turn, activates the MAPK/ERK pathway. These findings underline the relevance of iron in normal neuronal function.


Subject(s)
Animals , Rats , Extracellular Signal-Regulated MAP Kinases/drug effects , Iron/pharmacology , Neuroblastoma/enzymology , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Blotting, Western , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , /metabolism , Phosphorylation/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...