Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Horm Metab Res ; 55(8): 536-545, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37192655

RESUMEN

To evaluate safety and therapeutic effect along 12 months of allogenic adipose tissue-derived stromal/stem cells (ASCs) transplantation with cholecalciferol (VITD) in patients with recent-onset type 1 diabetes (T1D). Prospective, phase II, open trial, pilot study in which patients with recent onset T1D received ASCs (1xKgx106 cells) and VITD 2000UI/day for 12 months (group 1) and were compared to controls with standard insulin therapy (group 2). Adverse events, C-peptide area under the curve (CPAUC), insulin dose, HbA1c and frequency of FoxP3+ in CD4+ or CD8+ T-cells(flow cytometry) were evaluated at baseline(T0), after 3(T3), 6(T6) and 12 months(T12). Eleven patients completed follow up (7:group 1;4:group 2). Group 1 had lower insulin requirement at T3(0.24±0.18vs0.53±0.23UI/kg,p=0.04), T6(0.24±0.15vs0.66±0.33 UI/kg,p=0.04) and T12(0.39±0.15vs0.74±0.29 UI/Kg,p=0.04).HbA1c was lower at T6 (50.57±8.56vs72.25±10.34 mmol/mol,p=0.01), without differences at T12 (57.14±11.98 in group 1 vs. 73.5±14.57 mmol/min in group 2, p=0.16). CPAUC was not significantly different between groups at T0(p=0.07), higher in group 1 at T3(p=0.04) and T6(p=0.006), but similar at T12(p=0.23). IDAA1c was significantly lower in group 1 than group 2 at T3,T6 and T12 (p=0.006, 0.006 and 0.042, respectively). IDDA1c was inversely correlated to FoxP3 expression in CD4 and CD8+ T cells at T6 (p<0.001 and p=0.01, respectively). In group 1, one patient had recurrence of a benign teratoma that was surgically removed, not associated to the intervention. ASCs with VITD without immunosuppression were safe and associated lower insulin requirements, better glycemic control, and transient better pancreatic function in recent onset T1D, but the potential benefits were not sustained.


Asunto(s)
Diabetes Mellitus Tipo 1 , Humanos , Diabetes Mellitus Tipo 1/terapia , Colecalciferol/uso terapéutico , Hemoglobina Glucada , Proyectos Piloto , Estudios Prospectivos , Estudios de Seguimiento , Insulina/metabolismo , Tejido Adiposo/metabolismo , Suplementos Dietéticos , Células Madre/metabolismo , Factores de Transcripción Forkhead
2.
Artif Organs ; 45(6): 548-558, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33264436

RESUMEN

The new coronavirus (2019-nCoV) or the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was officially declared by the World Health Organization (WHO) as a pandemic in March 2020. To date, there are no specific antiviral drugs proven to be effective in treating SARS-CoV-2, requiring joint efforts from different research fronts to discover the best route of treatment. The first decisions in drug discovery are based on 2D cell culture using high-throughput screening. In this context, spheroids and organoids emerge as a reliable alternative. Both are scaffold-free 3D engineered constructs that recapitulate key cellular and molecular events of tissue physiology. Different studies have already shown their advantages as a model for different infectious diseases, including SARS-CoV-2 and for drug screening. The use of these 3D engineered tissues as an in vitro model can fill the gap between 2D cell culture and in vivo preclinical assays (animal models) as they could recapitulate the entire viral life cycle. The main objective of this review is to understand spheroid and organoid biology, highlighting their advantages and disadvantages, and how these scaffold-free engineered tissues can contribute to a better comprehension of viral infection by SARS-CoV-2 and to the development of in vitro high-throughput models for drug screening.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Organoides/fisiología , Esferoides Celulares/fisiología , Ingeniería de Tejidos/métodos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Humanos , Organoides/virología , SARS-CoV-2 , Esferoides Celulares/virología , Andamios del Tejido
3.
Front Cell Dev Biol ; 12: 1365671, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38344748

RESUMEN

[This corrects the article DOI: 10.3389/fcell.2022.1043117.].

4.
Artif Organs ; 37(12): 1068-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23865470

RESUMEN

The objective of our study was to investigate chondrogenesis potential of human adipose-derived mesenchymal stromal cells (MSCs), using as a positive control a human source of cartilage-derived progenitor cells (PCs). This source of PCs was recently described by our group and dwells on the surface of nasoseptal cartilage. Histological analysis using Safranin O staining and immunofluorescence for actin filaments and collagen type II was performed on three-dimensional (3D) pellet cultures. Cartilage PCs and adipose MSCs showed similarities in monolayer culture related to cell morphology and proliferation. Our 3D pellet cultures substantially reduced the actin stress and after 21 days under chondrogenic medium, we observed an increase in the pellet diameter for cartilage PCs (7.4%) and adipose MSCs (21.2%). Adipose-derived MSCs responded to chondrogenic stimulus, as seen by positive areas for collagen type II, but they were not able to recreate a mature extracellular matrix. Using semi-quantitative analysis, we observed a majority of Safranin O areas rising from blue (no stain) to orange (moderate staining) and no changes in fibroblastic morphology (P < 0.0001). For cartilage PCs, chondrogenic induction is responsible for morphological changes and a high percentage of matrix area/number of cells (P ≤ 0.0001), evaluated by computerized histomorphometry. Morphological analyses reveal that adipose-derived MSCs were not able to recreate a bioengineered cartilage. The cost of culture was reduced, as the cartilage PCs under growth-factor free medium exhibit a high score for cartilage formation compared with the induced adipose mesenchymal stromal cells (P = 0.0021). Using a pellet 3D culture, our cartilage PCs were able to produce a cartilage tissue in vitro, leading to the future development of bioengineered products.


Asunto(s)
Tejido Adiposo/metabolismo , Cartílago/metabolismo , Condrocitos/metabolismo , Condrogénesis , Células Madre Mesenquimatosas/metabolismo , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Citoesqueleto de Actina/metabolismo , Tejido Adiposo/citología , Adolescente , Adulto , Cartílago/citología , Proliferación Celular , Forma de la Célula , Células Cultivadas , Colágeno Tipo II/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Fenotipo , Factores de Tiempo , Adulto Joven
5.
Cells ; 12(12)2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37371053

RESUMEN

The immune and endocrine dysfunctions of white adipose tissue are a hallmark of metabolic disorders such as obesity and type 2 diabetes. In humans, white adipose tissue comprises distinct depots broadly distributed under the skin (hypodermis) and as internal depots (visceral). Depot-specific ASCs could account for visceral and subcutaneous adipose tissue properties, by regulating adipogenesis and immunomodulation. More importantly, visceral and subcutaneous depots account for distinct contributions to obesity and its metabolic comorbidities. Recently, distinct ASCs subpopulations were also described in subcutaneous adipose tissue. Interestingly, the superficial layer closer to the dermis shows hyperplastic and angiogenic capacities, whereas the deep layer is considered as having inflammatory properties similar to visceral. The aim of this focus review is to bring the light of recent discoveries into white adipose tissue heterogeneity together with the biology of distinct ASCs subpopulations and to explore adipose tissue 3D models revealing their advantages, disadvantages, and contributions to elucidate the role of ASCs in obesity development. Recent advances in adipose tissue organoids opened an avenue of possibilities to recreate the main cellular and molecular events of obesity leading to a deep understanding of this inflammatory disease besides contributing to drug discovery. Furthermore, 3D organ-on-a-chip will add reproducibility to these adipose tissue models contributing to their translation to the pharmaceutical industry.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Reproducibilidad de los Resultados , Diabetes Mellitus Tipo 2/metabolismo , Tejido Adiposo/metabolismo , Grasa Subcutánea , Obesidad/metabolismo
6.
Front Cell Dev Biol ; 10: 1043117, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36478741

RESUMEN

Medicine today faces the combined challenge of an increasing number of untreatable diseases and fewer drugs reaching the clinic. While pharmaceutical companies have increased the number of drugs in early development and entering phase I of clinical trials, fewer actually successfully pass phase III and launch into the market. In fact, only 1 out of every 9 drugs entering phase I will launch. In vitro preclinical tests are used to predict earlier and better the potential of new drugs and thus avoid expensive clinical trial phases. The most recent developments favor 3D cell culture and human stem cell biology. These 3D humanized models known as organoids better mimic the 3D tissue architecture and physiological cell behavior of healthy and disease models, but face critical issues in production such as small-scale batches, greater costs (when compared to monolayer cultures) and reproducibility. To become the gold standard and most relevant biological model for drug discovery and development, organoid technology needs to integrate biological culture processes with advanced microtechnologies, such as microphysiological systems based on microfluidics technology. Microphysiological systems, known as organ-on-a-chip, mimic physiological conditions better than conventional cell culture models since they can emulate perfusion, mechanical and other parameters crucial for tissue and organ physiology. In addition, they reduce labor cost and human error by supporting automated operation and reduce reagent use in miniaturized culture systems. There is thus a clear advantage in combining organoid culture with microsystems for drug development. The main objective of this review is to address the recent advances in organoids and microphysiological systems highlighting crucial technologies for reaching a synergistic strategy, including bioprinting.

7.
J Vis Exp ; (181)2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35435900

RESUMEN

Adipose-derived stromal/stem cells (ASCs) are a subpopulation of cells found in the stromal vascular fraction of human subcutaneous adipose tissue recognized as a classical source of mesenchymal stromal/stem cells. Many studies have been published with ASCs for scaffold-based tissue engineering approaches, which mainly explored the behavior of these cells after their seeding on bioactive scaffolds. However, scaffold-free approaches are emerging to engineer tissues in vitro and in vivo, mainly by using spheroids, to overcome the limitations of scaffold-based approaches. Spheroids are 3D microtissues formed by the self-assembly process. They can better mimic the architecture and microenvironment of native tissues, mainly due to the magnification of cell-to-cell and cell-to-extracellular matrix interactions. Recently, spheroids are mainly being explored as disease models, drug screening studies, and building blocks for 3D bioprinting. However, for 3D bioprinting approaches, numerous spheroids, homogeneous in size and shape, are necessary to biofabricate complex tissue and organ models. In addition, when spheroids are produced automatically, there is little chance for microbiological contamination, increasing the reproducibility of the method. The large-scale production of spheroids is considered the first mandatory step for developing a biofabrication line, which continues in the 3D bioprinting process and finishes in the full maturation of the tissue construct in bioreactors. However, the number of studies that explored the large-scale ASC spheroid production are still scarce, together with the number of studies that used ASC spheroids as building blocks for 3D bioprinting. Therefore, this article aims to show the large-scale production of ASC spheroids using a non-adhesive micromolded hydrogel technique spreading ASC spheroids as building blocks for 3D bioprinting approaches.


Asunto(s)
Bioimpresión , Tejido Adiposo/metabolismo , Bioimpresión/métodos , Humanos , Reproducibilidad de los Resultados , Esferoides Celulares , Células Madre , Ingeniería de Tejidos/métodos
8.
Front Bioeng Biotechnol ; 9: 682498, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34239860

RESUMEN

Cancer is considered one of the most predominant diseases in the world and one of the principal causes of mortality per year. The cellular and molecular mechanisms involved in the development and establishment of solid tumors can be defined as tumorigenesis. Recent technological advances in the 3D cell culture field have enabled the recapitulation of tumorigenesis in vitro, including the complexity of stromal microenvironment. The establishment of these 3D solid tumor models has a crucial role in personalized medicine and drug discovery. Recently, spheroids and organoids are being largely explored as 3D solid tumor models for recreating tumorigenesis in vitro. In spheroids, the solid tumor can be recreated from cancer cells, cancer stem cells, stromal and immune cell lineages. Organoids must be derived from tumor biopsies, including cancer and cancer stem cells. Both models are considered as a suitable model for drug assessment and high-throughput screening. The main advantages of 3D bioprinting are its ability to engineer complex and controllable 3D tissue models in a higher resolution. Although 3D bioprinting represents a promising technology, main challenges need to be addressed to improve the results in cancer research. The aim of this review is to explore (1) the principal cell components and extracellular matrix composition of solid tumor microenvironment; (2) the recapitulation of tumorigenesis in vitro using spheroids and organoids as 3D culture models; and (3) the opportunities, challenges, and applications of 3D bioprinting in this area.

9.
Tissue Eng Part A ; 27(5-6): 311-327, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-30734654

RESUMEN

A stabilized cartilage construct without signs of hypertrophy in chondrocytes is still a challenge. Suspensions of adipose stem/stromal cells (ASCs) and cartilage progenitor cells (CPCs) were seeded into micromolded nonadhesive hydrogel to produce spheroids (scaffold- and serum-free method) characterized by size, immunohistochemistry, fusion, and biomechanical properties. After cell dissociation, they were characterized for mesenchymal cell surface markers, cell viability, and quantitative real-time polymerase chain reaction. Both targeted and nontargeted (shotgun mass spectrometry) analyses were conducted on the culture supernatants. Induced ASC spheroids (ø = 350 µm) showed high cell viability and CD73 downregulation contrasting to CD90. The transforming growth factor (TGF)-ß3/TGF-ß1 ratio and SOX9 increased (p < 0.05), whereas interleukin (IL)-6, IL-8, RUNX2, and ALPL decreased. Induced ASC spheroids were able to completely fuse and showed a higher force required to compression at day 14 (p < 0.0001). Strong collagen type II in situ was associated with gradual decrease of collagen type X and a lower COLXA1 gene expression at day 14 compared with day 7 (p = 0.0352). The comparison of the secretome content of induced and non-induced ASCs and CPCs identified 138 proteins directly relevant to chondrogenesis of 704 proteins in total. Although collagen X was absent, thrombospondin-1 (TSP-1), described as antiangiogenic and antihypertrophic, and cartilage oligomeric matrix protein (COMP), a biomarker of chondrogenesis, were upregulated in induced ASC spheroids. Our scaffold- and serum-free method mimics stable cartilage acting as a tool for biomarker discovery and for regenerative medicine protocols. Impact Statement Promising adult stem cell sources for cartilage regeneration include adipose stem/stromal cells (ASCs) from subcutaneous adipose tissue. Our main objective was the development of a reproducible and easy-to-handle scaffold- and serum-free method to obtain stable cartilage from induced ASC spheroids. In addition to targeted protein profiling and biomechanical analysis, we provide the first characterization of the secretome composition for ASC spheroids, providing a useful tool to monitor in vitro chondrogenesis and a noninvasive quality control of tissue-engineered constructs. Furthermore, our secretome analysis revealed a potential novel biomarker-thrombospondin-1 (TSP-1), known by its antiangiogenic properties and recently described as an antihypertrophic protein.


Asunto(s)
Cartílago , Células Madre Mesenquimatosas , Tejido Adiposo , Diferenciación Celular , Células Cultivadas , Condrocitos , Condrogénesis , Humanos , Trombospondina 1 , Ingeniería de Tejidos
10.
Arch Endocrinol Metab ; 65(3): 342-351, 2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33939911

RESUMEN

OBJECTIVE: Adipose tissue-derived stromal/stem cells (ASCs) and vitamin D have immunomodulatory actions that could be useful for type 1 diabetes (T1D). We aimed in this study to investigate the safety and efficacy of ASCs + daily cholecalciferol (VIT D) for 6 months in patients with recent-onset T1D. METHODS: In this prospective, dual-center, open trial, patients with recent onset T1D received one dose of allogenic ASC (1 × 106 cells/kg) and cholecalciferol 2,000 UI/day for 6 months (group 1). They were compared to patients who received chol-ecalciferol (group 2) and standard treatment (group 3). Adverse events were recorded; C-peptide (CP), insulin dose and HbA1c were measured at baseline (T0), after 3 (T3) and 6 months (T6). RESULTS: In group 1 (n = 7), adverse events included transient headache (all), mild local reactions (all), tachycardia (n = 4), abdominal cramps (n = 1), thrombophlebitis (n = 4), scotomas (n = 2), and central retinal vein occlusion at T3 (n = 1, resolution at T6). Group 1 had an increase in basal CP (p = 0.018; mean: 40.41+/-40.79 %), without changes in stimulated CP after mixed meal (p = 0.62), from T0 to T6. Basal CP remained stable in groups 2 and 3 (p = 0.58 and p = 0.116, respectively). Group 1 had small insulin requirements (0.31+/- 0.26 UI/kg) without changes at T6 (p = 0.44) and HbA1c decline (p = 0.01). At T6, all patients (100%; n = 7) in group 1 were in honeymoon vs 75% (n = 3/4) and 50% (n = 3/6) in groups 2 and 3, p = 0.01. CONCLUSION: Allogenic ASC + VIT D without immunosuppression was safe and might have a role in the preservation of ß-cells in patients with recent-onset T1D. ClinicalTrials.gov: NCT03920397.


Asunto(s)
Colecalciferol/uso terapéutico , Diabetes Mellitus Tipo 1 , Trasplante de Células Madre Mesenquimatosas , Células Madre/citología , Tejido Adiposo/citología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Humanos , Proyectos Piloto , Estudios Prospectivos
11.
Brain Res ; 1747: 147026, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32750328

RESUMEN

Despite the regenerative potential of the Peripheral Nervous System (PNS), injuries with loss of a nerve segment make the functional recovery a challenge. This work aimed to investigate the effects of the association of biodegradable conduits of poly (lactic acid) (PLA) with human adipose-derived stem cells (hADSCs) on the regeneration of the sciatic nerve. C57BL / 6 male mice were submitted to sciatic nerve transection followed by tubulization with PLA conduit. Animals were allocated in two groups: the first received an injection of DMEM inside the conduit (DMEM) and the second received hADSCs inside it (hADSC). Sensory and motor functions were assessed by the pinprick test and electroneuromiography, respectively. To assess neuronal survival the retrograde tracer fluorogold was injected into the sciatic nerve distally to the lesion site. One week after that, animals were sacrificed, tissues harvested and processed for morphological evaluation. After eight weeks, all animals showed sensory recovery in the pinprick test and there was no significant difference between the two groups. The amplitude of the compound muscle action potential was higher in the hADSCs group. The number of myelinated nerve fibers, muscle cells and motor plates was higher in the hADSC group. There was also greater survival of sensory and motor neurons in the hADSC animals. These results suggest that the association of PLA conduit and cell therapy with hADSCs leads to a better functional and morphological recovery after sciatic nerve transection.


Asunto(s)
Tejido Adiposo/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático/lesiones , Células Madre/citología , Animales , Supervivencia Celular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Traumatismos de los Nervios Periféricos/fisiopatología , Poliésteres , Recuperación de la Función/fisiología , Nervio Ciático/fisiología
12.
Cytotherapy ; 11(6): 706-15, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19878057

RESUMEN

BACKGROUND AIMS: Since initial methods were developed for isolating cells from adipose tissue, little has been done to improve mesenchymal stromal cell (MSC) yield. The aim of the present study was to isolate a population of MSC from lipoaspirate samples without tissue digestion and to assess the possibility of cryopreserving the freshly isolated cells. METHODS: A population of MSC was isolated from 13 patients' lipoaspirate samples by mechanical dissociation. Mechanically processed lipoaspirate adipose tissue (MPLA) cells were characterized after in vitro cell expansion by morphologic analysis, expression of MSC surface markers and differentiation assays. RESULTS: Mechanical dissociation yielded a large quantity of adherent MSC both after standard and vibro-assisted liposuction. Preservation of lipoaspirate samples at 4 degrees C for 1 or 2 days until the mechanical procedure did not change the MPLA cell content. It was possible to store freshly isolated MPLA cells by cryopreservation without loss of the MSC population. Adherent MPLA cells were negative for CD45 and CD31 and positive for CD34, CD105, CD44 and CD90. They also showed adipogenic, osteogenic and chondrogenic potentials similar to MSC populations from other sources as already described in the literature. CONCLUSIONS: MSC can be isolated from human lipoaspirate samples by the mechanical procedure described in this study with a significant reduction in time and cost. Together with cryopreservation of freshly isolated MPLA cells, this has made it easier to harvest and store MSC for therapeutic applications such as soft-tissue augmentation and tissue engineering.


Asunto(s)
Tejido Adiposo/citología , Separación Celular/métodos , Células Madre Mesenquimatosas/citología , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular , Criopreservación , Endoglina , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Superficie Celular/metabolismo , Antígenos Thy-1/metabolismo
13.
Cytotechnology ; 70(6): 1655-1669, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30386942

RESUMEN

Stem cell tissue constructs are likely to come into contact with silver-based nanoparticles-such as silver chloride nanoparticles (AgCl-NPs)-used as microbicidals at the implant site or in cosmetics. However, the effect of silver-based nanoparticles on 3D cell cultures with potential for tissue engineering has received little attention. Here, we examined the effect of sub-lethal doses (5, 10 and 25 µg/mL, for 1, 7 and 21 days) of AgCl-NPs produced by 'green' bacterial-based synthesis on spheroid 3D cultures of human adipose tissue stem cells (ASCs). Light microscopy analysis revealed that the shape and diameter of ASC spheroids remained largely unchanged after AgCl-NP treatment. Flow cytometry analysis with 7-AAD and 2',7'-dichlorofluorescein diacetate revealed no statistically significant differences in cell death but showed an increase of ROS levels for the untreated group and significant differences for the groups treated with 5 and 10 µg/mL at day 7 (p = 0.0395, p = 0.0266, respectively). Electron microscopy analysis showed limited cell damage in the periphery of AgCl-NP-treated spheroids. However, treatment with AgCl-NP had statistically significant effects on the secretion of IL-6, IL-8, IL-1ß and IL-10 by spheroids, at specific treatment periods and concentrations, and particularly for IL-6, IL-8 and IL-1ß. TGF-ß1 and -ß2 secretion also changed significantly throughout the treatment period. Our results indicate that, despite having little effect on cell viability and morphology, sub-lethal AgCL-NP doses modulate ROS production at day 7 for the groups treated with 5 and 10 µg/mL and also modulate the secretory profile of ASC spheroids. Thus, the use of skin implants or products containing Ag-NPs may promote long-term disturbances in subcutaneous adipose tissue homeostasis.

14.
Stem Cells Int ; 2017: 7053465, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29527227

RESUMEN

The scaffold-free tissue engineering using spheroids is pointed out as an approach for optimizing the delivery system of cartilage construct. In this study, we aimed to evaluate the micromolded nonadhesive hydrogel (MicroTissues®) for spheroid compaction (2-day culture) and spontaneous chondrogenesis (21-day culture) using cartilage progenitors cells (CPCs) from human nasal septum without chondrogenic stimulus. CPC spheroids showed diameter stability (486 µm ± 65), high percentage of viable cells (88.1 ± 2.1), and low percentage of apoptotic cells (2.3%). After spheroid compaction, the synthesis of TGF-ß1, TGF-ß2, and TGF-ß3 was significantly higher compared to monolayer (p < 0.005). Biomechanical assay revealed that the maximum forces applied to spheroids after chondrogenesis were 2.6 times higher than for those cultured for 2 days. After spontaneous chondrogenesis, CPC spheroids were entirely positive for N-cadherin, collagen type II and type VI, and aggrecan and chondroitin sulfate. Comparing to monolayer, the expression of SOX5 and SOX6 genes analyzed by qPCR was significantly upregulated (p < 0.01). Finally, we observed the capacity of CPC spheroids starting to fuse. To the best of our knowledge, this is the first time in the scientific literature that human CPC spheroids were formed by micromolded nonadhesive hydrogel, achieving a successful scaffold-free cartilage engineering without chondrogenic stimulus (low cost).

15.
PLoS One ; 11(11): e0166073, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27829016

RESUMEN

Adipose stem cells (ASCs) spheroids show enhanced regenerative effects compared to single cells. Also, spheroids have been recently introduced as building blocks in directed self-assembly strategy. Recent efforts aim to improve long-term cell retention and integration by the use of microencapsulation delivery systems that can rapidly integrate in the implantation site. Interlockable solid synthetic microscaffolds, so called lockyballs, were recently designed with hooks and loops to enhance cell retention and integration at the implantation site as well as to support spheroids aggregation after transplantation. Here we present an efficient methodology for human ASCs spheroids biofabrication and lockyballs cellularization using micro-molded non-adhesive agarose hydrogel. Lockyballs were produced using two-photon polymerization with an estimated mechanical strength. The Young's modulus was calculated at level 0.1362 +/-0.009 MPa. Interlocking in vitro test demonstrates high level of loading induced interlockability of fabricated lockyballs. Diameter measurements and elongation coefficient calculation revealed that human ASCs spheroids biofabricated in resections of micro-molded non-adhesive hydrogel had a more regular size distribution and shape than spheroids biofabricated in hanging drops. Cellularization of lockyballs using human ASCs spheroids did not alter the level of cells viability (p > 0,999) and gene fold expression for SOX-9 and RUNX2 (p > 0,195). The biofabrication of ASCs spheroids into lockyballs represents an innovative strategy in regenerative medicine, which combines solid scaffold-based and directed self-assembly approaches, fostering opportunities for rapid in situ biofabrication of 3D building-blocks.


Asunto(s)
Tejido Adiposo/citología , Esferoides Celulares/trasplante , Células Madre/citología , Andamios del Tejido/química , Adolescente , Adulto , Técnicas de Cultivo de Célula , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Módulo de Elasticidad , Femenino , Expresión Génica , Humanos , Hidrogeles/química , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Persona de Mediana Edad , Medicina Regenerativa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9/genética , Sefarosa/química , Esferoides Celulares/química , Esferoides Celulares/citología , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Células Madre/ultraestructura , Ingeniería de Tejidos/métodos , Adulto Joven
16.
Arch. endocrinol. metab. (Online) ; 65(3): 342-351, May-June 2021. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1285166

RESUMEN

ABSTRACT Objective: Adipose tissue-derived stromal/stem cells (ASCs) and vitamin D have immunomodulatory actions that could be useful for type 1 diabetes (T1D). We aimed in this study to investigate the safety and efficacy of ASCs + daily cholecalciferol (VIT D) for 6 months in patients with recent-onset T1D. Materials and methods: In this prospective, dual-center, open trial, patients with recent onset T1D received one dose of allogenic ASC (1 x 106 cells/kg) and cholecalciferol 2,000 UI/day for 6 months (group 1). They were compared to patients who received chol-ecalciferol (group 2) and standard treatment (group 3). Adverse events were recorded; C-peptide (CP), insulin dose and HbA1c were measured at baseline (T0), after 3 (T3) and 6 months (T6). Results: In group 1 (n = 7), adverse events included transient headache (all), mild local reactions (all), tachycardia (n = 4), abdominal cramps (n = 1), thrombophlebitis (n = 4), scotomas (n = 2), and central retinal vein occlusion at T3 (n = 1, resolution at T6). Group 1 had an increase in basal CP (p = 0.018; mean: 40.41+/-40.79 %), without changes in stimulated CP after mixed meal (p = 0.62), from T0 to T6. Basal CP remained stable in groups 2 and 3 (p = 0.58 and p = 0.116, respectively). Group 1 had small insulin requirements (0.31+/- 0.26 UI/kg) without changes at T6 (p = 0.44) and HbA1c decline (p = 0.01). At T6, all patients (100%; n = 7) in group 1 were in honeymoon vs 75% (n = 3/4) and 50% (n = 3/6) in groups 2 and 3, p = 0.01. Conclusions: Allogenic ASC + VIT D without immunosuppression was safe and might have a role in the preservation of β-cells in patients with recent-onset T1D. ClinicalTrials.gov: NCT03920397.


Asunto(s)
Humanos , Células Madre/citología , Colecalciferol/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Proyectos Piloto , Tejido Adiposo/citología , Estudios Prospectivos
17.
World J Stem Cells ; 7(1): 165-73, 2015 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-25621116

RESUMEN

The discovery that adipose tissue represents an interesting source of multipotent stem cells has led to many studies exploring the clinical potential of these cells in cell-based therapies. Recent advances in understanding the secretory capacity of adipose tissue and the role of adipokines in the development of obesity and associated disorders have added a new dimension to the study of adipose tissue biology in normal and diseased states. Subcutaneous adipose tissue forms the interface between the clinical application of regenerative medicine and the establishment of the pathological condition of obesity. These two facets of adipose tissue should be understood as potentially related phenomena. Because of the functional characteristics of adipose stem cells, these cells represent a fundamental tool for understanding how these two facets are interconnected and could be important for therapeutic applications. In fact, adipose tissue stem cells have multiple functions in obesity related to adipogenic, angiogenic and secretory capacities. In addition, we have also previously described a predominance of larger blood vessels and an adipogenic memory in the subcutaneous adipose tissue after massive weight loss subsequent to bariatric surgery (ex-obese patients). Understanding the reversibility of the behavior of adipose stem cells in obeses and in weight loss is relevant to both physiological studies and the potential use of these cells in regenerative medicine.

18.
Stem Cell Res Ther ; 6: 72, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25884374

RESUMEN

INTRODUCTION: Subcutaneous adipose tissue is an interesting source of autologous stem cells with a fundamental role in the pathophysiology of obesity, metabolic syndromes and insulin resistance. We hypothesize that obesity could alter the stromal-vascular fraction (SVF) and adipose stem cell (ASCs) functions, which could compromise its regenerative behavior. Furthermore, we aimed to evaluate whether ASCs derived from post bariatric surgery ex-obese women maintain their functions in a similar fashion as do those from individuals who have never been obese. METHODS: The SVF of subcutaneous adipose tissue from control (n = 6, body mass index - BMI - 27.5 ± 0.5 kg/m(2)), obese (n = 12, BMI 46.2 ± 5.1 kg/m(2)) and post bariatric surgery ex-obese (n = 7, initial BMI 47.8 ± 1.3 kg/m(2); final BMI 28.1 ± 1.1 kg/m(2)) women were isolated and evaluated by flow cytometry. ASCs were tested for lipid accumulation by perilipin, adipose differentiation-related protein (ADRP) and Oil Red O staining after adipogenic stimulus. The cytokines secreted by the ASCs and after lipid accumulation induction were also evaluated. RESULTS: The subcutaneous adipose tissue of obese and post bariatric surgery ex-obese women was enriched in pericytes (p = 0.0345). The number of supra-adventitial cells was not altered in the obese patients, but it was highly enriched in the post bariatric surgery ex-obese women (p = 0.0099). The ASCs of the post bariatric surgery ex-obese patients secreted more MCP-1 (monocyte chemoattractant protein-1; p = 0.0078). After lipid accumulation induction, the ASCs of the patients in all groups secreted less IL-6 than the ASCs with no adipogenic stimulus (p < 0.0001). Obese ASCs with lipid accumulation secreted the highest amount of IL-6 (p < 0.001) whereas the ASCs from the controls secreted the highest amount of adiponectin (p < 0.0001). The ASCs from the post bariatric surgery ex-obese patients showed the highest levels of lipid accumulation whereas those from the obese women had the lowest levels (p < 0.0001). CONCLUSIONS: SVF content and ASC behavior are altered in the subcutaneous adipose tissue of morbid obese women; these changes are not completely restored after bariatric surgery-induced weight loss. The cellular alterations described in this study could affect the regenerative effects of adipose stem cells. Further investigations are required to avoid jeopardizing the development of autologous stem cell-based therapies.


Asunto(s)
Obesidad Mórbida/patología , Células Madre/metabolismo , Grasa Subcutánea/citología , Adipogénesis , Adiponectina/metabolismo , Adulto , Adventicia/citología , Adventicia/metabolismo , Cirugía Bariátrica , Índice de Masa Corporal , Proteínas Portadoras/metabolismo , Quimiocina CCL2/metabolismo , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Pericitos/citología , Pericitos/metabolismo , Perilipina-1 , Perilipina-2 , Fosfoproteínas/metabolismo , Células Madre/citología
19.
Stem Cell Res ; 8(2): 292-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22099383

RESUMEN

In cartilaginous tissues, perichondrium cambium layer may be the source of new cartilage. Human nasal septal perichondrium is considered to be a homogeneous structure in which some authors do not recognize the perichondrium internal zone or the cambium layer as a layer distinct from adjacent cartilage surface. In the present study, we isolated a chondrogenic cell population from human nasal septal cartilage surface zone. Nasoseptal chondrogenic cells were positive for surface markers described for mesenchymal stem cells, with exception of CD146, a perivascular cell marker, which is consistent with their avascular niche in cartilage. Although only Sox-9 was constitutively expressed, they also revealed osteogenic and chondrogenic, but not adipogenic, potentials in vitro, suggesting a more restricted lineage potential compared to mesenchymal stem cells. Interestingly, even in absence of chondrogenic growth factors in the pellet culture system, nasoseptal chondrogenic cells had a capacity to synthesize sulfated glycosaminoglycans, large amounts of collagen type II and to a lesser extent collagen type I. The spontaneous chondrogenic potential of this population of cells indicates that they may be a possible source for cartilage tissue engineering. Besides, the pellet culture system using nasoseptal chondrogenic cells may also be a model for studies of chondrogenesis.


Asunto(s)
Cartílago/fisiología , Separación Celular/métodos , Condrocitos/citología , Condrogénesis , Tabique Nasal/citología , Ingeniería de Tejidos/métodos , Adipogénesis , Adulto , Técnicas de Cultivo de Célula , Linaje de la Célula , Condrocitos/ultraestructura , Humanos , Tabique Nasal/ultraestructura , Osteogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA