Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Aesthetic Plast Surg ; 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789811

RESUMEN

BACKGROUND: Autologous adipose tissue is an ideal material for soft tissue filling and transplantation; however, high volumes of fat absorption over time lead to a relatively low overall survival percentage. The survival and differentiation of adipose-derived stem cells (ADSCs) in the transplanted microenvironment might improve adipose graft survival. Adipocytes have been reported to affect ADSC activation. However, its underlying mechanisms remain unclear. METHODS: Human ADSCs were incubated in a culture medium supplemented with hypoxic or normoxic conditioned culture medium (CM) derived from human adipocytes. Neuronal Pentraxin 1 (NPTX1) was overexpressed or knocked down in human adipocytes using an overexpression vector (NPTX1 OE) or small interfering RNA (siRNA) transfection, respectively. ADSC differentiation and paracrine secretion were assessed. Nude mice were implanted with human adipocytes and ADSCs. The adipose tissue was subsequently evaluated by histological analysis. RESULTS: CM from hypoxic-stimulated human adipocytes significantly facilitated the differentiation ability and paracrine levels of ADSCs. NPTX1 was significantly up-regulated in human adipocytes exposed to hypoxic conditions. In vitro, CM derived from hypoxia-stimulated human adipocytes or NPTX1-overexpressing human adipocytes exposed to normoxia promoted ADSC differentiation and paracrine; after silencing NPTX1, the facilitating effects of hypoxia-treated human adipocytes on ADSC activation were eliminated. Similarly, in vivo, the NPTX1 OE + normoxia-CM group saw improved histological morphology and fat integrity, less fibrosis and inflammation, and increased vessel numbers compared with the OE NC + normoxia-CM group; the adipocyte grafts of the si-NC + hypoxia-CM group yielded the most improved histological morphology, fat integrity, and the most vessel numbers. However, these enhancements of ADSC activation and adipose graft survival were partially abolished by NPTX1 knockdown in human adipocytes. CONCLUSION: NPTX1 might mediate the facilitating effects of hypoxia-stimulated human adipocytes on ADSC activation, thereby improving adipose tissue survival rate after autologous fat transplantation and the effectiveness of autologous fat transplantation through promoting ADSC activation. LEVEL OF EVIDENCE III: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .

2.
Exp Dermatol ; 31(12): 1837-1852, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35102608

RESUMEN

Stem cell-mediated therapies in combination with biomaterial and growth factor-based approaches in regenerative medicine are rapidly evolving with increasing application beyond the dermatologic field. Adipose-derived stem cells (ADSCs) are the more frequently used adult stem cells due to their abundance and easy access. In the case of volumetric defects, adipose tissue can take the shape of defects, restoring the volume and enhancing the regeneration of receiving tissue. When regenerative purposes prevail on volume restoration, the stromal vascular fraction (SVF) rich in staminal cells, purified mesenchymal stem cells (MSCs) or their cell-free derivatives grafting are favoured. The therapeutic efficacy of acellular approaches is explained by the fact that a significant part of the natural propensity of stem cells to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines and extracellular matrix components. Therefore, the secretome's ability to modulate multiple targets simultaneously demonstrated preclinical and clinical efficacy in reversing pathological mechanisms of complex conditions such atopic dermatitis (AD), vitiligo, psoriasis, acne and Lichen sclerosus (LS), non-resolving wounds and alopecia. This review analysing both in vivo and in vitro models gives an overview of the clinical relevance of adipose tissue-derivatives such as autologous fat graft, stromal vascular fraction, purified stem cells and secretome for skin disorders application. Finally, we highlighted the major disease-specific limitations and the future perspective in this field.


Asunto(s)
Dermatología , Células Madre Mesenquimatosas , Tejido Adiposo , Medicina Regenerativa , Células Madre
3.
Connect Tissue Res ; 62(3): 325-336, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32151168

RESUMEN

Purpose: Adipose-derived stem cells (ADSCs) are ideal for cell-based therapies to support bone regeneration. It is vital to understand the critical genes and molecular mechanisms involved in the functional regulation of ADSCs for enhancing bone regeneration. In the present study, we investigated the Gremlin 1 (GREM1) effect on ADSCs osteogenic differentiation and senescence.Materials and methods: The in vitro ADSCs osteogenic differentiation potential was evaluated by determining alkaline phosphatase (ALP) activity, mineralization ability, and the expression of osteogenic markers. Cell senescence is determined by SA-ß-gal staining, telomerase assay, and the expression of aging markers.Results: GREM1 overexpression in ADSCs reduced ALP activity and mineralization, inhibited the expression of osteogenic related genes OCN, OPN, DSPP, DMP1, and BSP, and key transcription factors, RUNX2 and OSX. GREM1 knockdown in ADSCs enhanced ALP activity and mineralization, promoted the expression of OCN, OPN, DSPP, DMP1, BSP, RUNX2, and OSX. GREM1 overexpression in ADSCs reduced the percent SA-ß-Gal positive cells, P16 and P53 expressions, and increased telomerase activity. GREM1 knockdown in ADSCs increased the percentage of SA-ß-Gal positive cells, P16 and P53 expressions, and reduced telomerase activity. Furthermore, GREM1 reduced the mRNA expression levels of BMP2, BMP6, and BMP7.Conclusions: In summary, our findings suggested that GREM1 inhibited ADSCs senescence and osteogenic differentiation and antagonized BMP transcription.


Asunto(s)
Osteogénesis , Telomerasa , Diferenciación Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Osteogénesis/genética , Células Madre , Telomerasa/genética , Proteína p53 Supresora de Tumor
4.
Exp Cell Res ; 388(2): 111816, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31923426

RESUMEN

Lymphedema is a chronic progressive disease ultimately resulting in severe, disfiguring swelling and permanent changes of the affected tissues. Presently, there is no causal treatment approach of lymphedema. Therefore, most therapies are purely symptomatic. However, the recent use of stem cell-based therapies has offered new prospects for alternative treatment options. The present study was performed to investigate the effects of human adipose-derived stem cells (ADSCs) on human dermal lymphatic endothelial cells (HDLECs) in terms of basic in vitro lymphangiogenic assays (WST-8 assay, scratch assay, transmigration assay, sprouting assay, tube formation assay). The influence of ADSC-conditioned medium (ADSC-CM) on HDLECs was compared to recombinant VEGF-C, bFGF and HGF. Further ADSC-CM was characterized by protein microarray and enzyme-linked immunosorbent assay (ELISA). Although key-lymphangiogenic growth factors - like VEGF-C - could only be detected in low concentrations within the conditioned medium (CM), HDLECs were potently stimulated to proliferate, migrate and to form tube like structures by ADSC-CM. Despite concentrations more than hundredfold higher than those found in the conditioned medium, stimulation with recombinant VEGF-C, bFGF and HGF was still weaker compared to ADSC-CM. These results highlight the effectiveness of growth factors secreted by ADSC to stimulate HDLEC, potentially providing a promising new therapeutic approach for the treatment of lymphedema.


Asunto(s)
Proliferación Celular , Dermis/citología , Células Endoteliales/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfangiogénesis , Células Madre Mesenquimatosas/citología , Movimiento Celular , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Dermis/efectos de los fármacos , Dermis/metabolismo , Células Endoteliales/metabolismo , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo
5.
Aesthetic Plast Surg ; 45(4): 1853-1859, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33770217

RESUMEN

OBJECTIVE: In the present study, we aimed to investigate the survival of stromal vascular fraction-gelatin (SVF-gel) grafts and determine whether co-transplantation of SVF-gel and platelet-rich fibrin (PRF) improves long-term maintenance of fat volume (Wei et al. in Oncotarget 8:68542-68556, 2017) in a rabbit model. METHODS: SVF-gel was transplanted into the ears of 12 rabbits with (experimental group) or without PRF (control group). Transplantation retention was evaluated based on weight, histology, and immunohistochemistry. RESULTS: In the 2nd and 4th weeks, the volume of fat was larger in the experimental group than in the control group. In the 6th week, the absorption of fat was noticeable in both groups, and there was no significant difference in the fat survival rate between the two groups (experimental group: 1.051 ± 0.144 and control group: 0.789 ± 0.232, P > 0.05). HE staining results: At week 2, adipocytes were observed in the experimental group and tended to mature over time. These adipocytes also exhibited an ordered arrangement. Adipocytes with abnormal morphology appeared in the control group in the 4th week. At different weeks, there were more inflammatory cells and fibroblasts in the experimental group than in the control group, and they were arranged in an ordered fashion. Immunohistochemical results: More brown areas were observed in the experimental group than in the control group, and the morphology and distribution of adipocytes in the experimental group were better than those in the control group. The distribution of fibrocytes was also more regular in the experimental group than in the control group. CONCLUSION: SVF-gel cannot maintain long-term filling in rabbit ears. The addition of PRF has no influence, although PRF can induce SVF-gel to transform into adipocytes, and the anti-inflammatory effect is noticeable in the early period following the procedure. Co-transplantation also helped to ensure orderly arrangement of fibrin. There were no "volume preservation differences in this experimental model" perhaps there are differences if other models/methodology are employed. No Level Assigned This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine Ratings, please refer to Table of Contents or online Instructions to Authors - www.springer.com/00266 .


Asunto(s)
Fibrina Rica en Plaquetas , Trasplantes , Adipocitos , Tejido Adiposo , Animales , Gelatina , Conejos , Células del Estroma
6.
Aesthetic Plast Surg ; 45(6): 2939-2947, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34085105

RESUMEN

BACKGROUND: Fat grafting has been extensively applied as natural filler and has been very promising in restoring volume loss. Lipografting has also been credited to reduce age-related skin changes due to the regenerative potential of adipose derived stem cells. Cell-mediated therapies in plastic surgery are rapidly evolving with growing applications. Nanofat, a bio-regenerative liquid suspension rich in stromal vascular fraction cells without viable adipocytes, has been described as an efficient cutaneous anti-aging therapy. We have published in 2013 a review entitled "stem cell facelift: between reality and fiction." Available clinical evidence at that time did not substantiate marketing and promotional claims of "stem cell facelift". The same year, the report about nanofat was published demonstrating striking clinical outcome. The current literature search is aimed at reviewing any evidence that has emerged since then regarding clinical efficacy of this modality. METHODS: A thorough PICO tool-based comprehensive literature search of PubMed database for "the efficacy of nanofat cell-mediated anti-aging therapy" was conducted with a time frame from 2013 till present. RESULTS: Despite apparent increasing popularity of stem cell rejuvenation, well-controlled clinical studies about this modality are surprisingly very scarce. Only seven papers published after 2013 were identified and were included in this review CONCLUSION: Though considered to be a safe procedure, and despite documented histologic improvement and striking clinical outcome in some reports, available evidence can hardly support clinical improvement of skin quality. Before cell-mediated aesthetic rejuvenation applications can be routinely undertaken, more robust evidence with well-defined primary outcome end points and objective outcome measures is required. LEVEL OF EVIDENCE: IV.


Asunto(s)
Ritidoplastia , Tejido Adiposo , Rejuvenecimiento , Células Madre , Fracción Vascular Estromal
7.
J Cell Physiol ; 235(11): 8736-8746, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32324266

RESUMEN

Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells that their therapeutic effects in various diseases make them an interesting tool in cell therapy. In the current study, we aimed to overexpress interferon-ß (IFN-ß) and leukemia inhibitory factor (LIF) cytokines in human ADSCs to evaluate the impact of this overexpression on human ADSCs properties. Here, we designed a construct containing IFN-ß and LIF and then, transduced human adipose-derived stem cells (hADSCs) by this construct via a lentiviral vector (PCDH-513B). We assessed the ability of long-term expression of the transgene in transduced cells by western blot analysis and enzyme-linked immunosorbent assay techniques on Days 15, 45, and 75 after transduction. For the evaluation of stem cell properties, flow cytometry and differentiation assays were performed. Finally, the MTT assay was done to assess the proliferation of transduced cells compares to controls. Our results showed high-efficiency transduction with highest expression rates on Day 75 after transduction which were 70 pg/ml for IFN-ß and 77.9 pg/ml for LIF in comparison with 25.60 pg/ml and 27.63 pg/ml, respectively, in untransduced cells (p = .0001). Also, transduced cells expressed a high level of ADSCs surface markers and successfully differentiated into adipocytes, chondrocytes, neural cells, and osteocytes besides the preservation rate of proliferation near untreated cells (p = .88). All in all, we successfully constructed an hADSC population stably overexpressed IFN-ß and LIF cytokines. Considering the IFN-ß and LIF anti-inflammatory and neuroprotective effects as well as immune-regulatory properties of hADSCs, the obtained cells of this study could be subjected for further evaluations in experimental autoimmune encephalomyelitis mice model.


Asunto(s)
Adipocitos/metabolismo , Interferón beta/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Humanos , Osteocitos/metabolismo , Células Madre/metabolismo
8.
Cell Biol Int ; 44(4): 1046-1058, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31930610

RESUMEN

Mesenchymal stem cells (MSCs) exists low efficiency to trans-differentiate into other germinal layer cell types. One key issue is to discover the effect of important factor on MSCs differentiation abiltiy. In this study, we investigated the role and mechanism of epiregulin (EREG) on the osteogenic differentiation and neurogenic trans-differentiation in adipose-derived stem cells (ADSCs). We discovered that the depletion of EREG inhibited the osteogenic differentiation in vitro. And 25 ng/mL recombinant human epiregulin protein (rhEREG) effectively improved the osteogenic differentiation of EREG-depleted-ADSCs. Depletion of EREG promoted the formation of neural spheres, and increased the expressions of nestin, ßIII-tubulin, NeuroD, NCAM, TH, and NEF in ADSCs. Then, 25 ng/mL rhEREG significantly inhibited these neurogenic differentiation indicators. Inhibition of p38 MAPK, JNK, or Erk1/2 signaling pathway separately, blocked the rhEREG-enhanced osteogenic differentiation ability and the rhEREG-inhibited neurogenic trans-differentiation ability of ADSCs. In conclusions, EREG promoted the osteogenic differentiation and inhibited the neurogenic trans-differentiation potentials of ADSCs via MAPK signaling pathways.


Asunto(s)
Transdiferenciación Celular , Epirregulina/metabolismo , Neurogénesis , Osteogénesis , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Aesthetic Plast Surg ; 44(4): 1230-1240, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32766907

RESUMEN

In this article, we tried to take stock of the state of the art in ultrasonic-assisted lipoplasty focusing on its most recent technologic advances and in the newest clinical applications, such as overweight, obesity, breast surgery and regenerative medicine. Great space was devoted to the analysis and clarification of the most common myths and legends related to this intriguing technique.


Asunto(s)
Lipectomía , Estética , Humanos , Obesidad/cirugía , Ultrasonido
10.
J Cell Physiol ; 234(2): 1452-1460, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30076718

RESUMEN

Adipose-derived stem cells (ADSCs) can differentiate into neurons under particular conditions. It remains largely unknown whether this differentiation potential is affected by physical conditions such as obesity, which modulates the functions of adipose tissue. In this study, we determined the impact of either a 9-week high-fat diet (60% fat; HFD) or 9-week exercise training on the differentiation potential of ADSCs into neuron-like cells in male Wistar rats. Rats were randomly assigned to a normal diet-fed (ND-SED) group, HFD-fed (HFD-SED) group, or exercise-trained HFD-fed group (HFD-EX). After a 9-week intervention, ADSCs from all groups differentiated into neuron-like cells. Expression of neuronal marker proteins (nestin, ßIII-tubulin, and microtubule-associated protein 2 [MAP2]) and the average length of cell neurites were lower in cells from HFD-SED rats than in other groups. Instead, protein expression of COX IV and Cyt-c, the Bax/Bcl-2 and LC3-II/I ratio, and the malondialdehyde level in culture medium were higher in cells from HFD-SED rats. No significant difference between ND-SED and HFD-EX rats was observed, except for the average length of cell neurites in MAP2. Thus, HFD impaired the differentiation potential of ADSCs into neuron-like cells, which was accompanied by increases in apoptotic activity and oxidative stress. Importantly, exercise training ameliorated the HFD-induced impairment of neurogenesis in ADSCs. The adipose tissue microenvironment could influence the differentiation potential of ADSCs, a source of autologous stem cell therapy.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células-Madre Neurales/patología , Neurogénesis , Neuronas/patología , Estrés Oxidativo , Condicionamiento Físico Animal , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Proteínas Relacionadas con la Autofagia/metabolismo , Linaje de la Célula , Células Cultivadas , Microambiente Celular , Masculino , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Neuritas/patología , Neuronas/metabolismo , Ratas Wistar , Carrera
11.
J Cell Biochem ; 120(3): 4433-4443, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30362610

RESUMEN

Multiple studies demonstrated that early growth response factor 1 (EGR1) induces myocardial damage after acute myocardial infarction (AMI). Recent evidence indicates that microRNAs (miRNA) play an important role in exosome-mediated cardioprotection after AMI. Bioinformatics analysis has shown that miR-146a can regulate the expression of EGR1, so the aim of this study was to determine if miR-146a plays a role in exosome-mediated cardioprotection by regulation of EGR1 after AMI. Exosomes were isolated from wild- or miR-146a-modified adipose-derived stem cells (ADSCs), and the therapeutic effect of exosomes was assessed in an AMI model in rats and hypoxic-induced H9c2 model cells. The results showed that miR-146a containing exosomes had more effect than the exosome treatment group on the suppression of AMI-induced apoptosis, inflammatory response, and fibrosis in an AMI rat model. Both in vivo and in vitro experiments found that miR-146a interacted with the 3'-untranslated region of EGR1 and suppressed posttranscriptional EGR1 expression, which was confirmed by the luciferase reporter assay. We also found that suppressed EGR1 expression reversed AMI or hypoxia-induced TLR4/NFκB signal activation, which played an important role in the promotion of myocardial cell apoptosis, inflammatory response, and fibrosis. Taken together, these findings suggested that exosomes derived from miR-146a-modified ADSCs attenuated AMI-induced myocardial damage via downregulation of EGR1.


Asunto(s)
Regulación hacia Abajo/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Exosomas/metabolismo , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Infarto del Miocardio/metabolismo , Regiones no Traducidas 3'/genética , Animales , Apoptosis/genética , Hipoxia de la Célula , Línea Celular , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Masculino , Miocardio/metabolismo , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo , Transfección
12.
J Cell Physiol ; 233(12): 9458-9472, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29995982

RESUMEN

Craniofacial defects can cause morbidness. Adipose-derived stem cells (ADSCs) have shown great promise for osteogeneration and vascularization; therefore cocultures of differentiated ADSCs are explored to increase bone and vessel formation. In this study, ADSCs were induced into osteogenic ADSCs (os-ADSCs) and endothelial ADSCs (endo-ADSCs) cells, which were then cocultured in variable proportions (os-ADSCs/endo-ADSCs = 2:1, 1:1, 1:2). The os-ADSCs in a ratio of 1:1 expressed more ALP, RUNX2 and COL-I, whereas VEGF, vWF and CD31 were upregulated in the endo-ADSCs of this group. Next generation RNA sequencing (RNA-seq) was performed to evaluate the molecular mechanisms of cocultured ADSCs. The os-ADSCs and endo-ADSCs interacted with each other during osteogenic and angiogenic differentiation, especially at the ratio of 1:1, and were regulated by vascular-related genes, cell-mediated genes, bone-related genes and the transforming growth factor ß signaling pathway (TGF-ß), mitogen-activated protein kinase signaling pathway (MAPK) and wnt signaling pathway (Wnt). Angptl4, apoe, mmp3, bmp6, mmp13 and fgf18 were detected to be up-regulated, and cxcl12 and wnt5a were down-regulated. The results showed that the gene expression levels were consistent with that in RNA-seq. The cells were then seeded into self-assembling peptide RADA16-I scaffolds as cocultures (1:1) and monocultures (ADSCs, os-ADSCs, endo-ADSCs). The results showed that the cells of all groups grew and proliferated well on the scaffolds, and the cocultured group exhibited better osteogeneration and vascularization. In conclusion, cocultured os-ADSCs and endo-ADSCs at the ratio of 1:1 showed strong osteogenic and angiogenic differentiation. There is a great potential for osteogenesis and vascularization by 3D culturing cells in a 1:1 ratio in self-assembling peptide RADA16-I scaffolds, which requires evaluation for bone regeneration in vivo.


Asunto(s)
Tejido Adiposo/citología , Regeneración Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Péptidos/farmacología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fenotipo , Ratas Sprague-Dawley , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/ultraestructura , Andamios del Tejido/química
13.
J Cell Biochem ; 119(10): 8450-8459, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30011072

RESUMEN

To investigate the function of epiregulin (EREG) in the migration and chemotaxis ability of mesenchymal stem cells. Adipose-derived stem cells (ADSCs) were used in this investigation. Lentiviral EREG short hairpin RNA was applied to silence EREG expression in ADSCs. Human recombinant EREG protein (rhEREG) was used to perform a gain-of-function study. Scratch-simulated wound migration and transwell chemotaxis assays were used to examine the migration and chemotaxis capacity of ADSCs in vitro. Using a Western blot assay, the expressions of p38 mitogen-activated protein kinase (p38 MAPK), c-Jun N-terminal kinase (JNK), extracellular signal-regulated protein kinases 1 and 2 (Erk1/2), and protein kinase B were detected. Depletion of EREG caused by specific short hairpin RNA restrained the migration and chemotaxis ability of ADSCs and inhibited the expressions of phosphorylated p38 MAPK, JNK, and Erk1/2. rhEREG improved ADSCs migration and chemotaxis capacity, which was repressed by knockdown of EREG and rescued the expressions of phosphorylated p38 MAPK, JNK, and Erk1/2 impaired by silencing EREG. Furthermore, rhEREG-improved migration and chemotaxis ability in EREG-depleted-ADSCs was restricted by a specific inhibitor, SB203580, for blocking p38 MAPK signaling, PD98059 for blocking Erk1/2 signaling, or SP600125 for blocking JNK signaling in ADSCs separately. EREG promotes migration and chemotaxis ability of ADSCs through MAPK signaling pathways.


Asunto(s)
Movimiento Celular/fisiología , Quimiotaxis/fisiología , Epirregulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Análisis de Varianza , Células Cultivadas , Epirregulina/genética , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Int J Mol Sci ; 19(12)2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30567301

RESUMEN

Human adipose-derived stem cells (hADSCs) are multipotent mesenchymal cells that can differentiate into adipocytes, chondrocytes, and osteocytes. During osteoblastogenesis, the osteoprogenitor cells differentiate into mature osteoblasts and synthesize bone matrix components. Zinc finger protein 521 (ZNF521/Zfp521) is a transcription co-factor implicated in the regulation of hematopoietic, neural, and mesenchymal stem cells, where it has been shown to inhibit adipogenic differentiation. The present study is aimed at determining the effects of ZNF521 on the osteoblastic differentiation of hADSCs to clarify whether it can influence their osteogenic commitment. The enforced expression or silencing of ZNF521 in hADSCs was achieved by lentiviral vector transduction. Cells were cultured in a commercial osteogenic medium for up to 20 days. The ZNF521 enforced expression significantly reduced osteoblast development as assessed by the morphological and molecular criteria, resulting in reduced levels of collagen I, alkaline phosphatase, osterix, osteopontin, and calcium deposits. Conversely, ZNF521 silencing, in response to osteoblastic stimuli, induced a significant increase in early molecular markers of osteogenesis and, at later stages, a remarkable enhancement of matrix mineralization. Together with our previous findings, these results show that ZNF521 inhibits both adipocytic and osteoblastic maturation in hADSCs and suggest that its expression may contribute to maintaining the immature properties of hADSCs.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Osteoblastos/citología , Osteogénesis/genética , Adipocitos/citología , Tejido Adiposo , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Osteoblastos/metabolismo , Células Madre/citología , Células Madre/metabolismo
15.
Int J Mol Sci ; 19(12)2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30558283

RESUMEN

Currently, the most effective therapy for liver diseases is liver transplantation, but its use is limited by organ donor shortage, economic reasons, and the requirement for lifelong immunosuppression. Mesenchymal stem cell (MSC) transplantation represents a promising alternative for treating liver pathologies in both human and veterinary medicine. Interestingly, these pathologies appear with a common clinical and pathological profile in the human and canine species; as a consequence, dogs may be a spontaneous model for clinical investigations in humans. The aim of this work was to characterize canine adipose-derived MSCs (cADSCs) and compare them to their human counterpart (hADSCs) in order to support the application of the canine model in cell-based therapy of liver diseases. Both cADSCs and hADSCs were successfully isolated from adipose tissue samples. The two cell populations shared a common fibroblast-like morphology, expression of stemness surface markers, and proliferation rate. When examining multilineage differentiation abilities, cADSCs showed lower adipogenic potential and higher osteogenic differentiation than human cells. Both cell populations retained high viability when kept in PBS at controlled temperature and up to 72 h, indicating the possibility of short-term storage and transportation. In addition, we evaluated the efficacy of autologous ADSCs transplantation in dogs with liver diseases. All animals exhibited significantly improved liver function, as evidenced by lower liver biomarkers levels measured after cells transplantation and evaluation of cytological specimens. These beneficial effects seem to be related to the immunomodulatory properties of stem cells. We therefore believe that such an approach could be a starting point for translating the results to the human clinical practice in future.


Asunto(s)
Tejido Adiposo/citología , Hepatopatías/terapia , Trasplante de Células Madre , Células Madre/citología , Adipogénesis , Adulto , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Cultivadas , Perros , Humanos , Osteogénesis , Adulto Joven
16.
World J Urol ; 34(10): 1447-55, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26743672

RESUMEN

PURPOSE: To evaluate the effect and possible mechanism of suburethral tissue-engineered sling in an animal model of stress urinary incontinence (SUI). METHODS: Adipose-derived stem cells (ADSCs) were obtained from the adipose tissues of rats. The differentiation potential, proliferation, and viability of rat ADSCs were evaluated after infecting these cells with a lentiviral vector-encoding green fluorescent protein (GFP). Next, GFP transfected ADSCs were seeded on polyglycolic acid (PGA) fibers to construct the tissue-engineered sling with the induction of 5-azacytidine (5-Aza). Afterward, the tissue-engineered slings were transplanted into a rat model of SUI that was established by vaginal balloon dilatation method and bilateral ovariectomy. Histology and the leak point pressure measurements were performed at 2 months after tissue-engineered sling implantation. RESULTS: The ADSCs were found to be efficiently transfected with GFP, without any effects on proliferation, cell cycle and multi-differentiation potential. After been seeded on PGA fibers, ADSCs formed tissue-engineered slings in 4 weeks of induction culture. Two months after implantation, the mean leak point pressure (LPP) was significantly increased in sling-treated rats compared with the balloon-injured ovariectomized rats. Immunofluorescence assay showed that some of the GFP expressing cells stained positive for muscle-specific markers. CONCLUSIONS: The newly suburethral tissue-engineered sling restores LPP in the rat model of SUI, which could be an effective treatment in future SUI therapy.


Asunto(s)
Tejido Adiposo/citología , Ácido Poliglicólico/farmacología , Células Madre/citología , Cabestrillo Suburetral , Ingeniería de Tejidos/métodos , Incontinencia Urinaria de Esfuerzo/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Ratas , Ratas Sprague-Dawley
17.
Biotechnol Bioeng ; 112(8): 1683-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25727058

RESUMEN

Three-dimensional (3D) cell manipulation is available with the integration of microfluidic technology and rapid prototyping techniques. High-Fidelity (Hi-Fi) constructs hold enormous therapeutic potential for organ manufacturing and regenerative medicine. In the present paper we introduced a quasi-three-dimensional (Q3D) model with parallel biocompatible alginate/gelatin/fibrin hurdles. The behaviors of fluids and cells along the microfluidic channels with various widths were studied. Cells inside the newly designed microfluidic channels attached and grew well. Morphological changes of adipose-derived stem cells (ADSCs) in both two-dimensional (2D) and 3D milieu were found on the printed constructs. Endothelialization occurred with the co-cultures of ADSCs and hepatocytes. This study provides insights into the interactions among fluids, cells and biomaterials, the behaviors of fluids and cells along the microfluidic channels, and the applications of Q3D techniques.


Asunto(s)
Células Endoteliales/fisiología , Hepatocitos/fisiología , Microfluídica/métodos , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Alginatos , Diferenciación Celular , Técnicas de Cocultivo , Fibrina , Gelatina , Ácido Glucurónico , Ácidos Hexurónicos , Andamios del Tejido
18.
Gland Surg ; 13(1): 45-58, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38323233

RESUMEN

Background: Adipose-derived stem cells (ADSCs) are closely associated with the survival rate of transplanted fat in breast reconstruction after breast cancer surgery. Nevertheless, the intrinsic mechanisms regulating ADSCs adipogenic differentiation remain ambiguous. The aim of our study was to explore the relevant genes and pathways to elucidate the potential mechanisms of adipogenic differentiation in ADSCs. Methods: The Gene Expression Omnibus (GEO) dataset GSE61302 was downloaded and analyzed to identify differentially expressed genes (DEGs). Key genes and signaling pathways were obtained through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional and enrichment analysis. Protein-protein interaction (PPI) network and hub gene analyses were performed with the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database and Cytoscape software. Finally, the transcription levels of hub genes in the adipogenic differentiated group and undifferentiated group of ADSCs were compared via real-time quantitative polymerase chain reaction (RT-qPCR). Results: In total, 1,091 DEGs were identified through bioinformatics analysis of the adipogenic differentiated group and undifferentiated group. If was then found that the 10 downregulated key genes, CCNB1, NUSAP1, DLGAP5, TTK, CCNB2, KIF23, BUB1B, CDC20, CDCA8, and KIF11 may play important roles in the adipogenic differentiation of ADSCs. Subsequent in vitro experimental verification also revealed that the messenger RNA (mRNA) expression levels of cyclin B1 in adipogenic differentiated cells and undifferentiated cells were significantly different at the early stage (P<0.05), but there was no significant difference at the late stage (P>0.05). Conclusions: As a key gene, CCNB1 might be a potential biomarker in the adipogenic differentiation of ADSCs at the early stage.

19.
Artículo en Inglés | MEDLINE | ID: mdl-38357905

RESUMEN

OBJECTIVE: The aim of this study was to construct a multicompartment synchronous rotating bioreactor (MCSRB) for batch-production of homogenized adipose-derived stem cell (ADSC) microspheres and treat neurogenic erectile dysfunction (ED). METHODS: Firstly, an MCSRB was constructed using a centrifugal device and hinged trays. Secondly, influence factors (density, rotational speed) on the formation of ADSC-spheroids were explored. Finally, a neurogenic ED model was established to verify the effectiveness and safety of ADSC-spheroids for ED treatment. RESULTS: An MCSRB promoted ADSCs to gather microspheres, most of which were 90-130 µm in diameter. Supernatant from three-dimensional culture led to a significant increase in cytokine expression in ADSCs and migration rate in human umbilical vein endothelial cells (HUVECs) compared to control groups. The erectile function and pathological changes of the penis were improved in the ADSC-spheroids treatment group compared to the traditional ADSCs treatment group (p < 0.01). CONCLUSION: Efficient, batch, controlled and homogenized production of ADSC stem cell microspheres, and effective improvement of erectile dysfunction in neurogenic rats can be achieved using the MCSRB device.

20.
Artículo en Inglés | MEDLINE | ID: mdl-38751673

RESUMEN

Background: Autologous fat transfer (AFT) is gaining popularity in breast surgery, offering a natural-looking and minimally invasive approach for augmentation, reconstruction, and contouring. However, concerns about its impact on breast cancer necessitate an understanding of the interplay between transplanted adipose-derived stem cells (ADSCs) and the breast tissue microenvironment. Renowned for regeneration, ADSCs raise questions about their role in cancer promotion. This systematic review delves into the complex relationship between AFT and breast cancer, exploring how ADSCs may influence development, growth, and metastasis. Methods: A systematic search of electronic databases, including PubMed, Embase, and BVS was conducted to identify relevant studies. The search strategy employed a combination of keywords, including "breast augmentation", "fat grafting", "breast enhancement", "mammoplasty", "cancer", "neoplasm" and related terms. Two reviewers independently screened titles and abstracts. Full-text articles were then retrieved for further evaluation based on their potential contribution to the review objectives. Results: Two hundred and forty records were identified. Among these, 104 duplicates were removed, resulting in 136 reports available for title and abstract screening. Subsequently, 54 papers were deemed potentially eligible for inclusion, and all reports were retrieved. Conclusions: In vitro studies reveal ADSCs dual role in breast cancer, influencing proliferation, migration, and drug resistance through complex signaling pathways. Animal studies highlight distinct ADSC subpopulations impacting tumor growth via direct interactions and extracellular vesicle cargo. In vivo, ADSC-enriched fat grafting is generally safe, showing no increased cancer recurrence risk compared to other methods. Notably, cases of invasive breast carcinoma warrant special attention. ADSC-enriched fat grafts exhibit potential benefits in graft retention and survival rates. Despite promising evidence, further studies are needed to comprehensively understand the intricate relationship between ADSCs and breast cancer for optimized clinical applications and potential therapeutic innovations.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda