Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Appl Biomater Funct Mater ; 21: 22808000231211423, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38131345

RESUMO

There are scarce published data suggesting, that collagen extracted from fish skin may be an attractive alternative to mammalian-derived collagen for the in vitro cell cultures. In this study, we investigated proliferation potential and differentiation capability into osteogenic and adipogenic lineages of rat adipose-derived mesenchymal stem cells (rASCs) and human adipose-derived mesenchymal stem cells (hASCs) cultured on collagen extracted from silver carp and African sharptooth catfish skins, compared with commercially available mammalian collagen and collagen-free culture dishes. Our results revealed no significant differences between fish collagen and mammalian collagen in supporting cell viability and proliferation capacity. Fish-derived collagen is a cheap material derived from production waste, does not contain transmissible pathogens of mammalian origin, supports human cell cultures at comparable level to conventional collagen sources, and may be considered as the product of choice for the in vitro cell cultures.


Assuntos
Tecido Adiposo , Células-Tronco Mesenquimais , Humanos , Ratos , Animais , Diferenciação Celular , Adipogenia , Colágeno , Osteogênese , Células Cultivadas , Mamíferos
2.
Tissue Cell ; 78: 101899, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36030673

RESUMO

BACKGROUND: The development of tissue-engineered scaffolds with electrical properties is the primary motivation of novel regenerative medicine. Electroconductive scaffolds are designed to mimic the injured tissue environment's electrical properties and regulate cellular behavior - growth, proliferation, and differentiation - that could stimulate the injured nerve's regeneration. METHODS: We fabricated dedicated electroconductive scaffolds and customized an appropriate device with an external current supply to expose cells on the scaffold to electrical stimulation (ES). Next, we isolated rat adipose-derived stem cells (ASCs) and performed in vitro experiments that combine cells, an electroconductive scaffold, NGF (nerve growth factor), and ES (90 mV/mm, constant, for four days). Finally, we checked cellular activity as proliferation, viability, morphology, the neurogenic differentiation potential of ASCs, cell alignment, and karyotype. RESULTS: We observed that the electrical stimulation did not change the viability and chromosome stability of rat ASCs, but altered slightly proliferation compared to non-stimulated cells. The combined effect of a scaffold, NGF, and ES caused morphology changes and enhancement of ASCs neuronal differentiation as indicated in ßIII-tubulin expression, actin organization, and upregulation of neurogenic gene expression. CONCLUSIONS: We developed an electroconductive scaffold and customized device for in vitro study with many experimental variants. Based on our results, we presumed that the established study scheme - including an electroconductive scaffold, NGF and ES - is biocompatible and could guide ASCs to differentiate in neurogenic lineage, thus may be potentially applied in nerve injury regeneration.


Assuntos
Células-Tronco Mesenquimais , Nanofibras , Actinas/metabolismo , Tecido Adiposo , Animais , Diferenciação Celular , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Ratos , Alicerces Teciduais , Tubulina (Proteína)
3.
Int J Mol Sci ; 22(11)2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-34070436

RESUMO

The aim was to examine the efficiency of a scaffold made of poly (L-lactic acid)-co-poly(ϵ-caprolactone), collagen (COL), polyaniline (PANI), and enriched with adipose-derived stem cells (ASCs) as a nerve conduit in a rat model. P(LLA-CL)-COL-PANI scaffold was optimized and electrospun into a tubular-shaped structure. Adipose tissue from 10 Lewis rats was harvested for ASCs culture. A total of 28 inbred male Lewis rats underwent sciatic nerve transection and excision of a 10 mm nerve trunk fragment. In Group A, the nerve gap remained untouched; in Group B, an excised trunk was used as an autograft; in Group C, nerve stumps were secured with P(LLA-CL)-COL-PANI conduit; in Group D, P(LLA-CL)-COL-PANI conduit was enriched with ASCs. After 6 months of observation, rats were sacrificed. Gastrocnemius muscles and sciatic nerves were harvested for weight, histology analysis, and nerve fiber count analyses. Group A showed advanced atrophy of the muscle, and each intervention (B, C, D) prevented muscle mass decrease (p < 0.0001); however, ASCs addition decreased efficiency vs. autograft (p < 0.05). Nerve fiber count revealed a superior effect in the nerve fiber density observed in the groups with the use of conduit (D vs. B p < 0.0001, C vs. B p < 0.001). P(LLA-CL)-COL-PANI conduits with ASCs showed promising results in managing nerve gap by decreasing muscle atrophy.


Assuntos
Modelos Animais de Doenças , Células-Tronco Mesenquimais/metabolismo , Nanofibras/química , Regeneração Nervosa , Neurogênese , Traumatismos dos Nervos Periféricos/terapia , Nervo Isquiático/metabolismo , Alicerces Teciduais/química , Compostos de Anilina/química , Animais , Caproatos/química , Células Cultivadas , Colágeno/química , Imuno-Histoquímica , Lactonas/química , Masculino , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica de Varredura , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Nanofibras/ultraestrutura , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Poliésteres/química , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático/citologia , Nervo Isquiático/patologia , Transplante Autólogo
4.
J Vis Exp ; (146)2019 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-31081818

RESUMO

The P19 cell line derived from a mouse embryo-derived teratocarcinoma has the ability to differentiate into the three germ layers. In the presence of retinoic acid (RA), the suspension cultured P19 cell line is induced to differentiate into neurons. This phenomenon is extensively investigated as a neurogenesis model in vitro. Therefore, the P19 cell line is very useful for molecular and cellular studies associated with neurogenesis. However, protocols for neuronal differentiation of P19 cell line described in the literature are very complex. The method developed in this study are simple and will play a part in elucidating the molecular mechanisms in neurodevelopmental abnormalities and neurodegenerative diseases.


Assuntos
Células-Tronco de Carcinoma Embrionário/patologia , Neurogênese , Animais , Diferenciação Celular/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Neurogênese/efeitos dos fármacos , Tretinoína/farmacologia
5.
Adv Clin Exp Med ; 27(8): 1085-1090, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29989681

RESUMO

BACKGROUND: Progress in breast cancer surgery results in a decreased frequency of mastectomy, in the early phases of cancer replaced by breast conserving therapy (lumpectomy). Increased popularity of breast reconstruction by fat or adipose stem cells (ASC)-enriched fat transfer raised uncertainty about the possible risk of increased cancer recurrence. In vitro studies suggest that locally secreted cytokines and reconstructed local blood vessels may stimulate cancer expansion or cancer de novo induction from glandular tissue remaining after lumpectomy. OBJECTIVES: The purpose of the study was to evaluate the risk of cancer recurrence in breast cancer patients related to the stromal vascular fraction (SVF) augmentation during autologous fat grafting for breast reconstruction. MATERIAL AND METHODS: The tumor recurrence ratio in 56 patients having the breast reconstructed with autologous ASC (transplanted as the subpopulation present in SVF) was compared with the frequency of tumor recurrence in 252 matched patients treated in clinics without subsequent breast reconstruction. Adipose tissue was collected by the Coleman technique and split into 2 portions: one was used for breast reconstruction, the other was enzymatically digested, and isolated cells were used for the augmentation of fat implanted into the breast area. Cancer recurrence in the experimental and matched control group was evaluated following 3-year-long observation time, and the statistical significance of difference in cancer recurrence between the experimental and control group was evaluated. RESULTS: Cancer recurrence in the group of patients treated with ASC-enriched fat for breast reconstruction was 3.7% and did not differ significantly from the control group data (4.13%). No adverse effects of therapy were observed. CONCLUSIONS: Our study does not produce any data suggesting increased cancer risk following breast reconstruction after a mastectomy or a lumpectomy combined with local radiotherapy. It may be concluded that an autologous transplantation of fat augmented with ASC is a safe and efficient procedure. Longer observation time and the observation of larger numbers of patients would be useful for strengthening the conclusion.


Assuntos
Tecido Adiposo/transplante , Mamoplastia/efeitos adversos , Mamoplastia/métodos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Recidiva Local de Neoplasia/epidemiologia , Adulto , Idoso , Neoplasias da Mama/cirurgia , Feminino , Humanos , Células-Tronco Mesenquimais , Pessoa de Meia-Idade , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos
6.
Adv Clin Exp Med ; 27(5): 717-723, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29952148

RESUMO

Mesenchymal stromal cells (MSCs) are an excellent and easily accessible source of precursor cells that have applications in regenerative medicine. They can be obtained from almost any tissue; however, bone marrow, Wharton's jelly and adipose tissue are the most frequently used sources of MSCs. Increased interest in using MSCs in medical procedures has resulted in a pressing need to identify the genetic elements that can indicate the presence and the characteristics of MSCs. Genomic profiling enables the identification and characterization of MSCs as well as finding biomarkers and key molecules involved in all processes occurring in the cell. This knowledge is essential for developing a stem cell approach for tissue engineering and can improve the development of new clinical applications of MSCs. This review is an attempt to give an overview of key genetic markers indicating the main directions of MSC differentiation. The expression of these genes provides information about the direction and progress of differentiation and about interactions with the surrounding environment as well as specific molecular pathways that MSCs are involved in.


Assuntos
Diferenciação Celular/genética , Marcadores Genéticos , Células-Tronco Mesenquimais , Biomarcadores , Células Cultivadas , Engenharia Tecidual , Geleia de Wharton
7.
J Obstet Gynaecol Res ; 43(11): 1758-1768, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28707770

RESUMO

AIM: The study was conducted to investigate secretory activity and define the paracrine potential of mesenchymal stem cells from human umbilical cord and amniotic membrane (UC-MSCs and AM-MSCs, respectively). METHODS: UC-MSCs (n = 6) were obtained from tissue explants using an adherent method after two weeks of incubation. AM-MSCs (n = 6) were obtained by digestion with tripsin and collagenase. MSC phenotype was confirmed in vitro by performing flow cytometry, differentiation assays and vimentin staining. Supernatants were collected after 48 h culturing in serum-free conditions and the following concentrations were determined: epidermal growth factor (EGF), interleukin (IL)-6, IL-10, tumor necrosis factor-α, transforming growth factor-ß (TGF-ß), vascular endothelial growth factor-α (VEGF-α) and metalloproteinase (MMP) 1, 8 and 13, using multiplex supernatant cytokine assay. Data were compared with adipose tissue derived MSCs (AD-MSCs, n = 6). RESULTS: Both UC-MSC and AM-MSC populations were positively identified as MSCs by flow cytometry and differentiation potential into bone, cartilage and adipose tissue. Using a multiple cytokine detection assay, we proved that both UC-MSCs and AM-MSCs show high secretive capacity. However, the secretion profile differed between cells from various sources. UC-MSCs showed significantly higher production of TGF-ß and lower production of VEGF-α, compared to AD-MSCs (P = 0.004) and AM-MSCs (P = 0.039) and lower levels of EGF (P = 0005). AM-MSCs showed significantly lower levels of MMP-8 than UC-MSCs (P = 0.024); however, there was no difference in levels of released cytokines compared to AD-MSCs. CONCLUSION: AM-MSCs show similar IL production as AD-MSCs, while UC-MSCs have a significantly different profile, which suggests diverse biological potential of both cell types for immunomodulative and regenerative therapy.


Assuntos
Tecido Adiposo , Âmnio , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Cordão Umbilical , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Âmnio/citologia , Âmnio/imunologia , Âmnio/metabolismo , Humanos , Cordão Umbilical/citologia , Cordão Umbilical/imunologia , Cordão Umbilical/metabolismo
8.
Gynecol Obstet Invest ; 82(3): 267-275, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27627653

RESUMO

BACKGROUND/AIM: Mesenchymal stem cells (MSCs) are gaining rising interest in gynecology and obstetrics. MSCs immunomodulatory properties are suitable enough to reduce perinatal morbidity caused by inflammation in premature neonates. The aim of this study was to evaluate and compare the ability to inhibit allo-activated lymphocytes proliferation by MSCs derived from different sources: amniotic membrane (AM), umbilical cord (UC) and adipose tissue (AT). METHODS: MSCs were isolated from AM (n = 7) and UC (n = 6) and AT (n = 6) of healthy women. Cells were characterized by flow cytometry and differentiation assay. To evaluate the potential of fetal and adult MSCs to diminish immunological response, mixed lymphocytes reaction (MLR) was performed. RESULTS: Amnion and UC-derived cells displayed typical MSCs characteristics. Addition of MSCs to MLR significantly inhibited the proliferation of stimulated lymphocytes. The effect was observed regardless of the MSCs type used (p < 0.01 in all groups). Comparative analysis revealed no significant differences in this action between tested MSCs types. Additionally, no type of MSCs significantly stimulated allogeneic lymphocytes. CONCLUSION: The results prove the immunosuppressive capacities of fetal-derived MSCs in vitro. In the future, they may be potentially used to treat premature newborn as well as in immunomodulation in post-transplant therapy.


Assuntos
Aloenxertos/imunologia , Âmnio/citologia , Células-Tronco Mesenquimais/imunologia , Cordão Umbilical/citologia , Tecido Adiposo/citologia , Adulto , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Imunomodulação/imunologia , Recém-Nascido , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Gravidez
9.
Mediators Inflamm ; 2016: 5302120, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27761060

RESUMO

The clinical outcome of autologous adipose stem cell (ASC) treatment of patients with multiple sclerosis (MS) was investigated following one year of observation. Methods. The clinical and MRI outcomes of 16 ASC-treated patients with RRMS and SPMS are reported after a one-year follow-up period. Results. At 18 months of follow-up, some patients showed "enticing" improvements on some exploratory efficacy measures, although a significant benefit was not observed for any measure across the entire group. Neither the progression of disability nor relapses were observed in any cases. In four patients, we found new gadolinium+ (Gd+) lesions on MRI. Our results indicate that ASC therapy is safe and does not produce any substantial side effects. Disease progression-free survival (PFS) of 18 months was seen in all patients with RRMS and SPMS. In these patients, EDSS scores did not progress above baseline scores. Gd-enhancing lesions were observed in two cases with RRMS, but these patients did not exhibit changes in EDSS score. Conclusion. Intrathecal treatment with ASCs is an attractive form of therapy for patients with MS but should be reserved for cases with aggressive disease progression, for cases that are still in the inflammatory phase, and for the malignant form.


Assuntos
Tecido Adiposo/citologia , Esclerose Múltipla/patologia , Esclerose Múltipla/terapia , Células-Tronco/citologia , Adulto , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Transplante de Células-Tronco , Células-Tronco/fisiologia
10.
Stem Cells Int ; 2016: 5762916, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27698672

RESUMO

Although mesenchymal stem cells are used in numerous clinical trials, the safety of their application is still a matter of concern. We have analysed the clinical results of the autologous adipose-derived stem cell treatment (stromal vascular fraction (SVF) containing adipose-derived stem cells, endothelial progenitors, and blood mononuclear cells) for orthopedic (cartilage, bone, tendon, or combined joint injuries) and neurologic (multiple sclerosis) diseases. Methods of adipose tissue collection, cell isolation and purification, and resulting cell numbers, viability, and morphology were considered, and patient's age, sex, disease type, and method of cell administration (cell numbers per single application, treatment numbers and frequency, and methods of cell implantation) were analysed and searched for the unwanted clinical effects. Results of cellular therapy were compared retrospectively to those obtained with conventional medication without SVF application. SVF transplantation was always the accessory treatment of patients receiving "standard routine" therapies of their diseases. Clinical experiments were approved by the Bioethical Medical Committees supervising the centers where patients were hospitalised. The conclusion of the study is that none of the treated patients developed any serious adverse event, and autologous mesenchymal stem (stromal) cell clinical application is a safe procedure resulting in some beneficial clinical effects (not analysed in this study).

11.
Oncotarget ; 7(41): 67412-67424, 2016 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-27602952

RESUMO

The CEBPA gene is known to be mutated or abnormally expressed in several cancers. This is the first study assessing the clinical impact of CEBPA gene status and expression on the ovarian cancer outcome. The CEBPA gene sequence was analyzed in 118 ovarian cancer patients (44 platinum/cyclophosphamide (PC)-treated and 74 taxane/platinum (TP)-treated), both in tumors and blood samples, and in blood from 236 healthy women, using PCR-Sanger sequencing and Real-Time quantitative PCR (qPCR)-based genotyping methods, respectively. The CEBPA mRNA level was examined with Reverse Transcription quantitative PCR (RT-qPCR). The results were correlated to different clinicopathological parameters. Thirty of 118 (25.4%) tumors harbored the CEBPA synonymous c.690G>T polymorphism (rs34529039), that we showed to be related to up-regulation of CEBPA mRNA levels (p=0.0059). The presence of the polymorphism was significantly associated with poor prognosis (p=0.005) and poor response to the PC chemotherapy regimen (p=0.024). In accordance, elevated CEBPA mRNA levels negatively affected patient survival (p<0.001) and tumor response to the PC therapy (p=0.014). The rs34529039 SNP did not affect the risk of developing ovarian cancer. This is the first study providing evidence that the c.690G>T, p.(Thr230Thr) (rs34529039) polymorphism of the CEBPA gene, together with up-regulation of its mRNA expression, are negative factors worsening ovarian cancer outcome. Their adverse clinical effect depends on a therapeutic regimen used, which might make them potential prognostic and predictive biomarkers for response to DNA-damaging chemotherapy.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Neoplasias Ovarianas/genética , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Intervalo Livre de Doença , Feminino , Genótipo , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/mortalidade , Polimorfismo de Nucleotídeo Único , Prognóstico , Modelos de Riscos Proporcionais , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA