Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Artif Organs ; 48(7): 723-733, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38385713

RESUMEN

BACKGROUND: The SARS-CoV-2 pandemic has spurred an unparalleled scientific endeavor to elucidate the virus' structure, infection mechanisms, and pathogenesis. Two-dimensional culture systems have been instrumental in shedding light on numerous aspects of COVID-19. However, these in vitro systems lack the physiological complexity to comprehend the infection process and explore treatment options. Three-dimensional (3D) models have been proposed to fill the gap between 2D cultures and in vivo studies. Specifically, spheroids, composed of lung cell types, have been suggested for studying SARS-CoV-2 infection and serving as a drug screening platform. METHODS: 3D lung spheroids were prepared by coculturing human alveolar or bronchial epithelial cells with human lung stromal cells. The morphology, size, and ultrastructure of spheroids before and after SARS-CoV-2 infection were analyzed using optical and electron microscopy. Immunohistochemistry was used to detect spike protein and, thus, the virus presence in the spheroids. Multiplex analysis elucidated the cytokine release after virus infection. RESULTS: The spheroids were stable and kept their size and morphology after SARS-CoV-2 infection despite the presence of multivesicular bodies, endoplasmic reticulum rearrangement, tubular compartment-enclosed vesicles, and the accumulation of viral particles. The spheroid responded to the infection releasing IL-6 and IL-8 cytokines. CONCLUSION: This study demonstrates that coculture spheroids of epithelial and stromal cells can serve as a cost-effective infection model for the SARS-CoV-2 virus. We suggest using this 3D spheroid as a drug screening platform to explore new treatments related to the cytokines released during virus infection, especially for long COVID treatment.


Asunto(s)
COVID-19 , Evaluación Preclínica de Medicamentos , Pulmón , SARS-CoV-2 , Esferoides Celulares , Humanos , Esferoides Celulares/virología , COVID-19/virología , SARS-CoV-2/fisiología , Pulmón/virología , Pulmón/patología , Tratamiento Farmacológico de COVID-19 , Antivirales/farmacología , Antivirales/uso terapéutico , Técnicas de Cocultivo , Citocinas/metabolismo , Análisis Costo-Beneficio , Células Epiteliales/virología
2.
Artif Organs ; 45(10): 1208-1218, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34036603

RESUMEN

As an alternative to the classical tissue engineering approach, bottom-up tissue engineering emerges using building blocks in bioassembly technologies. Spheroids can be used as building blocks to reach a highly complex ordered tissue by their fusion (bioassembly), representing the foundation of biofabrication. In this study, we analyzed the biomechanical properties and the fusion capacity of human adipose stem/stromal cell (ASC) we spheroids during an in vitro model of hypertrophic cartilage established by our research group. Hypertrophic induced-ASC spheroids showed a statistically significant higher Young's modulus at weeks 2 (P < .001) and 3 (P < .0005) compared with non-induced. After fusion, non-induced and induced-ASC spheroids increased the contact area and decreased their pairs' total length. At weeks 3 and 5, induced-ASC spheroids did not fuse completely, and the cells migrate preferentially in the fusion contact region. Alizarin red O staining showed the highest intensity of staining in the fused induced-ASC spheroids at week 5, together with intense staining for collagen type I and osteocalcin. Transmission electron microscopy and element content analysis (X-ray Energy Dispersive Spectroscopy) revealed in the fused quartet at week 3 a crystal-like structure. Hypertrophic induction interferes with the intrinsic capacity of spheroids to fuse. The measurements of contact between spheroids during the fusion process, together with the change in viscoelastic profile to the plastic, will impact the establishment of bioassembly protocols using hypertrophic induced-ASC spheroids as building blocks in biofabrication.


Asunto(s)
Tejido Adiposo/citología , Cartílago/crecimiento & desarrollo , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Tejido Adiposo/fisiología , Fenómenos Biomecánicos , Cartílago/citología , Cartílago/ultraestructura , Células Cultivadas , Humanos , Hipertrofia , Células Madre Mesenquimatosas/fisiología , Microscopía Electrónica de Transmisión , Esferoides Celulares/fisiología , Esferoides Celulares/ultraestructura , Células del Estroma/fisiología
3.
Artif Organs ; 44(7): E288-E299, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31950507

RESUMEN

Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.


Asunto(s)
Cartílago/crecimiento & desarrollo , Condrogénesis/fisiología , Células Madre Mesenquimatosas/fisiología , Cultivo Primario de Células/métodos , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Cartílago/citología , Cartílago/ultraestructura , Diferenciación Celular/fisiología , Células Cultivadas , Colágeno Tipo X/metabolismo , Medio de Cultivo Libre de Suero , Matriz Extracelular/metabolismo , Humanos , Hipertrofia , Metaloproteinasa 13 de la Matriz/metabolismo , Microscopía Electrónica de Transmisión , Esferoides Celulares/fisiología , Esferoides Celulares/ultraestructura , Células del Estroma/fisiología
4.
Int J Mol Sci ; 19(5)2018 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-29693604

RESUMEN

Top-down tissue engineering aims to produce functional tissues using biomaterials as scaffolds, thus providing cues for cell proliferation and differentiation. Conversely, the bottom-up approach aims to precondition cells to form modular tissues units (building-blocks) represented by spheroids. In spheroid culture, adult stem cells are responsible for their extracellular matrix synthesis, re-creating structures at the tissue level. Spheroids from adult stem cells can be considered as organoids, since stem cells recapitulate differentiation pathways and also represent a promising approach for identifying new molecular targets (biomarkers) for diagnosis and therapy. Currently, spheroids can be used for scaffold-free (developmental engineering) or scaffold-based approaches. The scaffold promotes better spatial organization of individual spheroids and provides a defined geometry for their 3D assembly in larger and complex tissues. Furthermore, spheroids exhibit potent angiogenic and vasculogenic capacity and serve as efficient vascularization units in porous scaffolds for bone tissue engineering. An automated combinatorial approach that integrates spheroids into scaffolds is starting to be investigated for macro-scale tissue biofabrication.


Asunto(s)
Células Madre Adultas/citología , Huesos/citología , Cartílago/citología , Esferoides Celulares/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Células Madre Adultas/fisiología , Animales , Proliferación Celular , Humanos , Nanofibras/química , Esferoides Celulares/fisiología
5.
Tissue Eng Part A ; 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38062998

RESUMEN

Three-dimensional (3D) bioprinting, a promising advancement in tissue engineering technology, involves the robotic, layer-by-layer additive biofabrication of functional 3D tissue and organ constructs. This process utilizes biomaterials, typically hydrogels and living cells, following digital models. Traditional tissue engineering uses a classic triad of living cells, scaffolds, and physicochemical signals in bioreactors. A scaffold is a temporary, often biodegradable, support structure. Tissue engineering primarily falls into two categories: (i) scaffold based and (ii) scaffold free. The latter, scaffold-free 3D bioprinting, is gaining increasing popularity. Organ building blocks (OBB), capable of self-assembly and self-organization, such as tissue spheroids, organoids, and assembloids, have begun to be utilized in scaffold-free bioprinting. This article discusses the expanding range of OBB, presents the rapidly evolving collection of bioprinting and bioassembly methods using these OBB, and finally, outlines the advantages, challenges, and future perspectives of using OBB in organ printing.

6.
Front Cell Dev Biol ; 11: 1219218, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37842092

RESUMEN

Background: Adipose tissue engineering may provide 3D models for the understanding of diseases such as obesity and type II diabetes. Recently, distinct adipose stem/stromal cell (ASC) subpopulations were identified from subcutaneous adipose tissue (SAT): superficial (sSAT), deep (dSAT), and the superficial retinacula cutis (sRC). This study aimed to test these subpopulations ASCs in 3D spheroid culture induced for adipogenesis under a pro-inflammatory stimulus with lipopolysaccharide (LPS). Methods: The samples of abdominal human subcutaneous adipose tissue were obtained during plastic aesthetic surgery (Protocol 145/09). Results: ASC spheroids showed high response to adipogenic induction in sSAT. All ASC spheroids increased their capacity to lipolysis under LPS. However, spheroids from dSAT were higher than from sSAT (p = 0.0045) and sRC (p = 0.0005). Newly formed spheroids and spheroids under LPS stimulus from sSAT showed the highest levels of fatty acid-binding protein 4 (FABP4) and CCAAT/enhancer-binding protein-α (C/EBPα) mRNA expression compared with dSAT and sRC (p < 0.0001). ASC spheroids from sRC showed the highest synthesis of angiogenic cytokines such as vascular endothelial growth factor (VEGF) compared with dSAT (p < 0.0228). Under LPS stimulus, ASC spheroids from sRC showed the highest synthesis of pro-inflammatory cytokines such as IL-6 compared with dSAT (p < 0.0092). Conclusion: Distinct physiological properties of SAT can be recapitulated in ASC spheroids. In summary, the ASC spheroid from dSAT showed the greatest lipolytic capacity, from sSAT the greatest adipogenic induction, and sRC showed greater secretory capacity when compared to the dSAT. Together, all these capacities form a true mimicry of SAT and hold the potential to contribute for a deeper understanding of cellular and molecular mechanisms in healthy and unhealthy adipose tissue scenarios or in response to pharmacological interventions.

7.
J Funct Biomater ; 14(12)2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-38132809

RESUMEN

Bone critical-size defects and non-union fractures have no intrinsic capacity for self-healing. In this context, the emergence of bone engineering has allowed the development of functional alternatives. The aim of this study was to evaluate the capacity of ASC spheroids in bone regeneration using a synergic strategy with 3D-printed scaffolds made from poly (lactic acid) (PLA) and nanostructured hydroxyapatite doped with carbonate ions (CHA) in a rat model of cranial critical-size defect. In summary, a set of results suggests that ASC spheroidal constructs promoted bone regeneration. In vitro results showed that ASC spheroids were able to spread and interact with the 3D-printed scaffold, synthesizing crucial growth factors and cytokines for bone regeneration, such as VEGF. Histological results after 3 and 6 months of implantation showed the formation of new bone tissue in the PLA/CHA scaffolds that were seeded with ASC spheroids. In conclusion, the presence of ASC spheroids in the PLA/CHA 3D-printed scaffolds seems to successfully promote bone formation, which can be crucial for a significant clinical improvement in critical bone defect regeneration.

8.
Polymers (Basel) ; 14(23)2022 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-36501463

RESUMEN

The concept of "lockyballs" or interlockable mini-scaffolds fabricated by two-photon polymerization from biodegradable polymers for the encagement of tissue spheroids and their delivery into the desired location in the human body has been recently introduced. In order to improve control of delivery, positioning, and assembly of mini-scaffolds with tissue spheroids inside, they must be functionalized. This review describes the design, fabrication, and functionalization of mini-scaffolds as well as perspectives on their application in tissue engineering for precisely controlled cell and mini-tissue delivery and patterning. The development of functionalized mini-scaffolds advances the original concept of "lockyballs" and opens exciting new prospectives for mini-scaffolds' applications in tissue engineering and regenerative medicine and their eventual clinical translation.

9.
Stem Cell Res Ther ; 12(1): 480, 2021 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-34454629

RESUMEN

BACKGROUND: In human subcutaneous adipose tissue, the superficial fascia distinguishes superficial and deep microenvironments showing extensions called retinacula cutis. The superficial subcutaneous adipose tissue has been described as hyperplastic and the deep subcutaneous adipose tissue as inflammatory. However, few studies have described stromal-vascular fraction (SVF) content and adipose-derived stromal/stem cells (ASCs) behavior derived from superficial and deep subcutaneous adipose tissue. In this study, we analyzed a third conjunctive microenvironment: the retinacula cutis superficialis derived from superficial subcutaneous adipose tissue. METHODS: The samples of abdominal human subcutaneous adipose tissue were obtained during plastic aesthetic surgery in France (Declaration DC-2008-162) and Brazil (Protocol 145/09). RESULTS: The SVF content was characterized in situ by immunofluorescence and ex vivo by flow cytometry revealing a high content of pre-adipocytes rather in superficial subcutaneous adipose tissue microenvironment. Adipogenic assays revealed higher percentage of lipid accumulation area in ASCs from superficial subcutaneous adipose tissue compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001). The high adipogenic potential of superficial subcutaneous adipose tissue was corroborated by an up-regulation of adipocyte fatty acid-binding protein (FABP4) compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001) and of C/EBPα (CCAAT/enhancer-binding protein alpha) compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001) microenvironments. Curiously, ASCs from retinacula cutis superficialis showed a higher level of adiponectin receptor gene compared with superficial subcutaneous adipose tissue (p = 0.0409), widely known as an anti-inflammatory hormone. Non-induced ASCs from retinacula cutis superficialis showed higher secretion of human vascular endothelial growth factor (VEGF), compared with superficial (p = 0.0485) and deep (p = 0.0112) subcutaneous adipose tissue and with adipogenic-induced ASCs from superficial (p = 0.0175) and deep (p = 0.0328) subcutaneous adipose tissue. Furthermore, ASCs from retinacula cutis superficialis showed higher secretion of Chemokine (C-C motif) ligand 5 (CCL5) compared with non-induced (p = 0.0029) and induced (p = 0.0089) superficial subcutaneous adipose tissue. CONCLUSIONS: This study highlights the contribution to ASCs from retinacula cutis superficialis in their angiogenic property previously described for the whole superficial subcutaneous adipose tissue besides supporting its adipogenic potential for superficial subcutaneous adipose tissue.


Asunto(s)
Tejido Subcutáneo , Factor A de Crecimiento Endotelial Vascular , Adipogénesis , Humanos , Grasa Subcutánea , Grasa Subcutánea Abdominal
10.
3D Print Addit Manuf ; 8(4): 227-237, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36654836

RESUMEN

Biocompatible scaffolds are porous matrices that are bone substitutes with great potential in tissue regeneration. For this, these scaffolds need to have bioactivity and biodegradability. From this perspective, 3D printing presents itself as one of the techniques with the greatest potential for scaffold manufacturing with porosity and established structure, based on 3D digital modeling. Thus, the objective of the present work was to produce 3D scaffolds from the poly (lactic acid) (PLA) and the nanostructured hydroxyapatite doped with carbonate ions (CHA). For this purpose, filaments were produced via fusion for the fused-filament 3D printing and used to produce scaffolds with 50% porosity in the cubic shape and 0/90°configuration. The dispersive energy spectroscopy and Fourier transform infrared spectroscopy (FTIR) analysis demonstrated the presence of CHA in the polymeric matrix, confirming the presence and incorporation into the composite. The thermogravimetric analysis made it possible to determine that the filler concentration incorporated in the matrix was very similar to the proposed percentage, indicating that there were no major losses in the process of obtaining the filaments. It can be assumed that the influence of CHA as a filler presents better mechanical properties up to a certain amount. The biological results point to a great potential for the application of PLA/CHA scaffolds in bone tissue engineering with effective cell adhesion, proliferation, biocompatibility, and no cytotoxicity effects.

11.
Aesthet Surg J ; 30(2): 249-55, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20442104

RESUMEN

BACKGROUND: Centrifugation is one of the preferred methods of fat processing. Although it has been promoted for nearly three decades to separate adipose tissue components before grafting, there remain many controversies regarding the results obtained with centrifuged adipose tissue. OBJECTIVES: The authors demonstrate the effects of centrifugation on the cellular components of aspirated fat. METHODS: Fat harvested from the lower abdomen of 10 female patients undergoing liposuction was divided in two equal parts, then processed by decantation or centrifugation and sent to the laboratory. Each processed lipoaspirate was analyzed histologically after hematoxylin and periodic acid-Schiff staining for the presence of intact adipocytes. It was then cultured and analyzed by multicolor flow cytometry for identification of adipose-derived mesenchymal stem cells. RESULTS: The middle layer of the centrifuged lipoaspirate, which is used by many surgeons, showed a great majority of altered adipocytes and very few mesenchymal stem cells in comparison with the decanted sample, which maintained the integrity of the adipocytes and showed a greater number of mesenchymal stem cells. The pellet observed as a fourth layer at the bottom of the centrifuged lipoaspirate showed the greatest concentration of endothelial cells and mesenchymal stem cells, which play a crucial role in the angiogenic and adipogenic effect of the grafted tissue. CONCLUSIONS: If centrifuged lipoaspirate is used, the pellet (rich in adipose-derived mesenchymal stem cells) and the middle layer should be employed to increase fat graft survival.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/trasplante , Centrifugación/métodos , Adipocitos , Adulto , Femenino , Citometría de Flujo , Humanos , Lipectomía , Células Madre Mesenquimatosas , Persona de Mediana Edad , Estudios Prospectivos , Coloración y Etiquetado
12.
World J Stem Cells ; 12(1): 1-7, 2020 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-32110271

RESUMEN

This article presents the stem and progenitor cells from subcutaneous adipose tissue, briefly comparing them with their bone marrow counterparts, and discussing their potential for use in regenerative medicine. Subcutaneous adipose tissue differs from other mesenchymal stromal/stem cells (MSCs) sources in that it contains a pre-adipocyte population that dwells in the adventitia of robust blood vessels. Pre-adipocytes are present both in the stromal-vascular fraction (SVF; freshly isolated cells) and in the adherent fraction of adipose stromal/stem cells (ASCs; in vitro expanded cells), and have an active role on the chronic inflammation environment established in obesity, likely due their monocytic-macrophage lineage identity. The SVF and ASCs have been explored in cell therapy protocols with relative success, given their paracrine and immunomodulatory effects. Importantly, the widely explored multipotentiality of ASCs has direct application in bone, cartilage and adipose tissue engineering. The aim of this editorial is to reinforce the peculiarities of the stem and progenitor cells from subcutaneous adipose tissue, revealing the spheroids as a recently described biotechnological tool for cell therapy and tissue engineering. Innovative cell culture techniques, in particular 3D scaffold-free cultures such as spheroids, are now available to increase the potential for regeneration and differentiation of mesenchymal lineages. Spheroids are being explored not only as a model for cell differentiation, but also as powerful 3D cell culture tools to maintain the stemness and expand the regenerative and differentiation capacities of mesenchymal cell lineages.

13.
World J Stem Cells ; 12(2): 110-122, 2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32184936

RESUMEN

Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by "bottom-up" approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.

14.
Front Immunol ; 11: 993, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582156

RESUMEN

Objective: To evaluate the short term safety and potential therapeutic effect of allogenic adipose tissue-derived stromal/stem cells (ASCs) + cholecalciferol in patients with recent-onset T1D. Methods: Prospective, phase II, open trial, pilot study in which patients with recent onset T1D received ASCs (1 × 106 cells/kg) and cholecalciferol 2000 UI/day for 3 months (group 1) and were compared to controls with standard insulin therapy (group 2). Adverse events, C-peptide (CP), insulin dose, HbA1c, time in range (TIR), glucose variability (continuous glucose monitoring) and frequency of CD4+FoxP3+ T-cells (flow cytometry) were evaluated at baseline (T0) and after 3 months (T3). Results: 13 patients were included (8: group 1; 5: group 2). Their mean age and disease duration were 26.7 ± 6.1 years and 2.9 ± 1.05 months. Adverse events were transient headache (n = 8), mild local reactions (n = 7), tachycardia (n = 4), abdominal cramps (n = 1), thrombophlebitis (n = 4), mild floaters (n = 2), central retinal vein occlusion (n = 1, complete resolution). At T3, group 1 had lower insulin requirement (0.22 ± 0.17 vs. 0.61±0.26IU/Kg; p = 0.01) and HbA1c (6.47 ± 0.86 vs. 7.48 ± 0.52%; p = 0.03) than group 2. In group 1, 2 patients became insulin free (for 4 and 8 weeks) and all were in honeymoon at T3 (vs. none in group 2; p = 0.01). CP variations did not differ between groups (-4.6 ± 29.1% vs. +2.3 ± 59.65%; p = 0.83). Conclusions: Allogenic ASCs + cholecalciferol without immunosuppression was associated with stability of CP and unanticipated mild transient adverse events in patients with recent onset T1D. ClinicalTrials.gov registration: NCT03920397.


Asunto(s)
Tejido Adiposo/citología , Colecalciferol/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Suplementos Dietéticos , Trasplante de Células Madre Mesenquimatosas , Vitaminas/uso terapéutico , Adolescente , Adulto , Biomarcadores/sangre , Glucemia/metabolismo , Brasil , Colecalciferol/efectos adversos , Terapia Combinada , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/diagnóstico , Suplementos Dietéticos/efectos adversos , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Proyectos Piloto , Estudios Prospectivos , Factores de Tiempo , Trasplante Homólogo , Resultado del Tratamiento , Vitaminas/efectos adversos , Adulto Joven
15.
Obes Surg ; 19(9): 1304-12, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19562421

RESUMEN

BACKGROUND: The normal function of white adipose tissue is disturbed in obesity. After weight loss that follows bariatric surgery, ex-obese patients undergo plastic surgery to remove residual tissues and it is not known whether their adipose tissue returns to its original state. The aim of this study was to compare the white adipose tissue composition of ex-obese with control patients with regard to blood vessels and resident mesenchymal stem cells (MSC). METHODS: Quantification of blood vessels was performed on histological sections of adipose tissue stained with hematoxylin and eosin and for von Willebrand antigen. MSC were induced to the adipogenic and osteogenic lineages by specific inductive culture media. Expression of PPARgamma2 was analyzed by reverse transcription polymerase chain reaction. RESULTS: Ex-obese adipose tissue showed a higher number (p = 0.0286) of small (107.3 +/- 22.0) and large (22.5 +/- 6.4) blood vessels, when compared to control patients (42.0 +/- 24.4 and 7.2 +/- 2.2, respectively) and they also occupied a larger area (control versus ex-obese, p = 0.0286). Adipose tissue MSC from both groups of patients expressed PPARgamma2 and were equally able to differentiate to the osteogenic lineage, but ex-obese MSC showed a higher adipogenic potential when induced in vitro (p < 0.05). CONCLUSIONS: The higher number of adipose tissue blood vessels in ex-obese patients explains the excessive bleeding observed during their plastic surgery. The presence of more committed cells to the adipogenic lineage may favor the easy weight regain that occurs in ex-obese patients. These results show that, after extensive weight loss, adipose tissue cell composition was not totally restored.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Obesidad/patología , Obesidad/fisiopatología , Grasa Subcutánea/irrigación sanguínea , Grasa Subcutánea/patología , Adulto , Índice de Masa Corporal , Estudios de Casos y Controles , Recuento de Células , Femenino , Humanos , Persona de Mediana Edad , Obesidad/cirugía , PPAR gamma/metabolismo , Grasa Subcutánea/metabolismo , Pérdida de Peso
16.
World J Stem Cells ; 11(3): 147-166, 2019 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-30949294

RESUMEN

The increasing prevalence of obesity is alarming because it is a risk factor for cardiovascular and metabolic diseases (such as type 2 diabetes). The occurrence of these comorbidities in obese patients can arise from white adipose tissue (WAT) dysfunctions, which affect metabolism, insulin sensitivity and promote local and systemic inflammation. In mammals, WAT depots at different anatomical locations (subcutaneous, preperitoneal and visceral) are highly heterogeneous in their morpho-phenotypic profiles and contribute differently to homeostasis and obesity development, depending on their ability to trigger and modulate WAT inflammation. This heterogeneity is likely due to the differential behavior of cells from each depot. Numerous studies suggest that adipose-derived stem/stromal cells (ASC; referred to as adipose progenitor cells, in vivo) with depot-specific gene expression profiles and adipogenic and immunomodulatory potentials are keys for the establishment of the morpho-functional heterogeneity between WAT depots, as well as for the development of depot-specific responses to metabolic challenges. In this review, we discuss depot-specific ASC properties and how they can contribute to the pathophysiology of obesity and metabolic disorders, to provide guidance for researchers and clinicians in the development of ASC-based therapeutic approaches.

17.
Toxicol In Vitro ; 57: 145-153, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30543842

RESUMEN

There are currently three in vitro methods adopted by the Organization for the Economic Co-operation and Development for testing chemicals based on the third key event of the skin sensitization adverse outcome pathway, the activation of dendritic cells. All of them use culture medium supplemented with fetal bovine serum (FBS), which brings technical disadvantages and animal welfare concerns. The objective of this study was to analyze the possibility of eliminating the use of FBS in the human Cell Line Activation Test (h-CLAT). After successful implementation of the h-CLAT using THP-1 cells cultured in FBS-containing medium, several attempts to adapt THP-1 cells to four different serum-free media were made. The best results were obtained with gradual adaptation to RPMI-1640 medium with HL-1™ Supplement and to X-VIVO™ 10. Adapted cells were cryopreserved and submitted to the reactivity check. After being approved, they were used in dose finding and proficiency assays. Despite minor adjustments in the original protocol, it was possible to correctly predict the sensitizing potential of the ten proficiency substances using THP-1 cells adapted to X-VIVO™ 10, which indicates that it is possible to eliminate the use of FBS in the h-CLAT, using a chemically defined medium.


Asunto(s)
Bioensayo/métodos , Haptenos/toxicidad , Pruebas de Irritación de la Piel/métodos , Alternativas a las Pruebas en Animales , Medios de Cultivo , Humanos , Células THP-1
18.
J Mol Endocrinol ; 63(1): 93-102, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067509

RESUMEN

Neuromedin B, a bombesin-like peptide, and its receptor, are expressed in white adipose tissue with undefined roles. Female mice with disruption of neuromedin B receptor (NB-R) exhibited partial resistance to diet-induced obesity leading to our hypothesis that NB-R is involved in adipogenesis. Here, we showed that adipose stem/stromal cells (ASC) from perigonadal fat of female NB-R-knockout mice, exposed to a differentiation protocol in vitro, accumulated less lipid (45%) than wild type, suggesting reduced capacity to differentiate under adipogenic input. To further explore mechanisms, preadipocytes 3T3-L1 cells were incubated in the presence of NB-R antagonist (PD168368) during the first 3 days in culture. Cells were analyzed in the end of the treatment (Day 3) and later when fully differentiated (Day 21). NB-R antagonist induced lower number of cells at day 3 and 21 (33-39%), reduced cell proliferation at day 3 (-53%) and reduced lipid accumulation at day 21 (-86%). The mRNA expressions of several adipocyte differentiation markers were importantly reduced at both days: Cebpb and Pparg and Fabp4, Plin-1 and Adipoq, and additionally Lep mRNA at day 21. The antagonist had no effect when incubated with mature 3T3-L1 adipocytes. Therefore, genetically disruption of NB-R in mice ASC or pharmacological antagonism of NB-R in 3T3-L1 cells impairs adipogenesis. The mechanisms suggested by results in 3T3-L1 cells involve reduction of cell proliferation and of early gene expressions, leading to decreased number of mature adipocytes. We speculate that NB-R antagonism may be useful to limit the increase in adiposity due to pre-adipocyte differentiation.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Receptores de Bombesina/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Indoles/farmacología , Ratones , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo , Perilipina-1/genética , Perilipina-1/metabolismo , Piridinas/farmacología , Receptores de Bombesina/antagonistas & inhibidores , Receptores de Bombesina/genética
19.
Front Biosci (Landmark Ed) ; 23(10): 1969-1986, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29772539

RESUMEN

Osteodegenerative disease and bone fractures lead to bone damage or loss, requiring new bone formation to replace the damaged tissues. Classical 'top-down' tissue engineering relies on seeding cell suspensions into biomaterial scaffolds, and then guiding cell fate by growth factors. However, complex tissue fabrication using this approach has important limitations. 'Bottom-up' tissue engineering has the potential to overcome the drawbacks of the top-down approach, by using 'building blocks' of cell spheroids for tissue biofabrication without a scaffold. Spheroids are 3D structures that resemble the physiological tissue microenvironment and can be produced in vitro by different methods. Spheroids of mesenchymal stem cells (MSC) and adipose stem cells (ASC) have regenerative properties. Here we review, the use of spheroids as 'building blocks' in the 3D bioprinting of large-scale bone tissue and as a promising alternative for the treatment of osteodegenerative diseases and in bone engineering, including endochondral ossification (or developmental engineering).


Asunto(s)
Huesos/fisiología , Esferoides Celulares/fisiología , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Huesos/citología , Diferenciación Celular/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Osteogénesis/fisiología , Esferoides Celulares/citología , Células Madre/citología , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA