Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(1)2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38203201

RESUMO

Wounds represent a common occurrence in human life. Consequently, scientific investigations are underway to advance wound healing methodologies, with a notable focus on dressings imbued with biologically active compounds capable of orchestrating the wound microenvironment through meticulously regulated release mechanisms. Among these bioactive agents are cytokines, which, when administered to the wound milieu without appropriate protection, undergo rapid loss of their functional attributes. Within the context of this research, we present a method for fabricating dressings enriched with G-CSF (granulocyte colony-stimulating factor) or GM-CSF (granulocyte-macrophage colony-stimulating factor), showcasing both biological activity and protracted release dynamics. Based on Ligasano, a commercial polyurethane foam dressing, and chitosan crosslinked with TPP (sodium tripolyphosphate), these dressings are noncytotoxic and enable cytokine incorporation. The recovery of cytokines from dressings varied based on the dressing preparation and storage techniques (without modification, drying, freeze-drying followed by storage at 4 °C or freeze-drying followed by storage at 24 °C) and cytokine type. Generally, drying reduced cytokine levels and their bioactivity, especially with G-CSF. The recovery of G-CSF from unmodified dressings was lower compared to GM-CSF (60% vs. 80%). In summary, our freeze-drying approach enables the storage of G-CSF or GM-CSF enriched dressings at 24 °C with minimal cytokine loss, preserving their biological activity and thus enhancing future clinical availability.


Assuntos
Quitosana , Surdez , Humanos , Citocinas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Granulócitos , Bandagens
2.
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
3.
J Liposome Res ; 27(2): 90-98, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28067107

RESUMO

Liposomes are used for encapsulation of the active compounds in different therapies, with the increasing frequency. The important areas of clinical applications of liposomes are cancer targeted treatment, antibiotic delivery or regenerative medicine. The liposomes can transfer both hydrophilic and hydrophobic compounds and have the lipid bilayer which imitates the cell membrane. Liposomes additionally may extend half-live period of drugs and protect them against the elimination in different ways, such as phagocytosis, enzymatic cleavage or exclusion by detoxification. The size and charge of liposomes play an important role in drug distribution and absorption into the cell. Limited data is available on the effects of liposomes on stem cells and progenitor cells. In this article, we examined the effect of charged conventional liposomes on growth of mesenchymal and blood stem cells isolated from umbilical cord. The data suggest a likelihood, that positively charged liposomes could impair stem cell growth and metabolism. Different methodological approaches allowed for the selection of negatively charged liposomes for further experiments, as the only type of liposomes which has the lowest cytotoxicity and does not affect hematopoietic cell proliferation.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Medicina Regenerativa , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Feminino , Humanos , Lipossomos/síntese química , Lipossomos/química , Lipossomos/farmacologia
4.
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
5.
Cells ; 13(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39272976

RESUMO

3D bioprinting has become a valuable tool for studying the biology of solid tumors, including glioblastoma multiforme (GBM). Our analysis of publicly available bulk RNA and single-cell sequencing data has allowed us to define the chemotactic profile of GBM tumors and identify the cell types that secrete particular chemokines in the GBM tumor microenvironment (TME). Our findings indicate that primary GBM tissues express multiple chemokines, whereas spherical monocultures of GBM cells significantly lose this diversity. Subsequently, the comparative analysis of GBM spherical monocultures vs. 3D-bioprinted multicultures of cells showed a restoration of chemokine profile diversity in 3D-bioprinted cultures. Furthermore, single-cell RNA-Seq analysis showed that cells of the perivascular niche (pericytes and endocytes) express multiple chemokines in the GBM TME. Next, we 3D-bioprinted cells from two glioblastoma cell lines, U-251 and DK-MG, alone and as co-cultures with mesenchymal stromal cells (representing cells of the perivascular niche) and assessed the chemokine secretome. The results clearly demonstrated that the interaction of tumors and mesenchymal cells leads to in a significant increase in the repertoire and levels of secreted chemokines under culture in 21% O2 and 1% O2. Our study indicates that cells of the perivascular niche may perform a substantial role in shaping the chemokine microenvironment in GBM tumors.


Assuntos
Quimiocinas , Técnicas de Cocultura , Glioblastoma , Células-Tronco Mesenquimais , Microambiente Tumoral , Humanos , Glioblastoma/patologia , Glioblastoma/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Quimiocinas/metabolismo , Linhagem Celular Tumoral , Impressão Tridimensional , Bioimpressão , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo
6.
Blood ; 117(16): e171-81, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21357765

RESUMO

Research applications and cell therapies involving genetically modified cells require reliable, standardized, and cost-effective methods for cell manipulation. We report a novel nanomagnetic method for integrated cell separation and gene delivery. Gene vectors associated with magnetic nanoparticles are used to transfect/transduce target cells while being passaged and separated through a high gradient magnetic field cell separation column. The integrated method yields excellent target cell purity and recovery. Nonviral and lentiviral magselectofection is efficient and highly specific for the target cell population as demonstrated with a K562/Jurkat T-cell mixture. Both mouse and human enriched hematopoietic stem cell pools were effectively transduced by lentiviral magselectofection, which did not affect the hematopoietic progenitor cell number determined by in vitro colony assays. Highly effective reconstitution of T and B lymphocytes was achieved by magselectofected murine wild-type lineage-negative Sca-1(+) cells transplanted into Il2rg(-/-) mice, stably expressing GFP in erythroid, myeloid, T-, and B-cell lineages. Furthermore, nonviral, lentiviral, and adenoviral magselectofection yielded high transfection/transduction efficiency in human umbilical cord mesenchymal stem cells and was fully compatible with their differentiation potential. Upscaling to a clinically approved automated cell separation device was feasible. Hence, once optimized, validated, and approved, the method may greatly facilitate the generation of genetically engineered cells for cell therapies.


Assuntos
Separação Celular/métodos , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Animais , Antígenos Ly/genética , Vetores Genéticos/química , Células-Tronco Hematopoéticas/metabolismo , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Células Jurkat , Células K562 , Magnetismo , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Nanopartículas/química , Transfecção
7.
Postepy Biochem ; 59(2): 187-97, 2013.
Artigo em Polonês | MEDLINE | ID: mdl-24044283

RESUMO

The multipotential progenitor cells called ,Mesenchymal Stem Cells" (MSC) are capable of differrentiation at least into bone, cartilage, and adipose tissues. The commonly recognized role of these cells is the formation of connective tissue which participates in formation of every organ. The progeny of MSC produces also the hematopoietic microenvironment, recently it have been documented that these cells are capable of the modulation of the immune system activities. MSC are isolated from the tissues of fetal origin (umbilical cord, cord blood, or placenta), or from several adult donor sites, in particular from bone marrow and adipose tissue which are most useful for practical purposes. The capability of multipotential differentiation, immunomodulation, and the regulation of the endogenous tissue repair are the reasons why mesenchymal stem cells are widely applied for regenerative medicine purposes.


Assuntos
Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Adulto , Osso e Ossos/citologia , Cartilagem/citologia , Diferenciação Celular , Feto/citologia , Humanos , Regeneração/fisiologia
8.
J Biomater Appl ; 38(4): 548-561, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37732423

RESUMO

The study aimed to evaluate an angiogenic effect of adipose-derived stem cells (ASCs) seeding and surgical prefabrication (placing a vascular pedicle inside the scaffold) on developed composite scaffolds made of poly-ε-caprolactone (PCL), ß-tricalcium phosphate (ß-TCP), and poly (lactic-co-glycolic acid) (PLGA) (PCL+ß-TCP+PLGA). Moreover, we aimed to compare our data with previously tested PCL scaffolds to assess whether the new material has better angiogenic properties. The study included 18 inbred male WAG rats. There were three scaffold groups (six animals each): with non-seeded PCL+ß-TCP+PLGA scaffolds, with PCL+ß-TCP+PLGA scaffolds seeded with ASCs and with PCL+ß-TCP+PLGA scaffolds seeded with ASCs and osteogenic-induced. Each rat was implanted with two scaffolds in the inguinal region (one prefabricated and one non-prefabricated). After 2 months from implantation, the scaffolds were explanted, and vessel density was determined by histopathological examination. Prefabricated ASC-seeded PCL+ß-TCP+PLGA scaffolds promoted greater vessel formation than non-seeded scaffolds (19.73 ± 5.46 vs 12.54 ± 0.81; p = .006) and those seeded with osteogenic-induced ASCs (19.73 ± 5.46 vs 11.87±2.21; p = .004). The developed composite scaffold promotes vessel formation more effectively than the previously described PCL scaffold.


Assuntos
Fosfatos de Cálcio , Alicerces Teciduais , Masculino , Ratos , Animais , Fosfatos de Cálcio/farmacologia , Adipócitos , Osteogênese , Células-Tronco
9.
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
10.
Mutagenesis ; 27(5): 551-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22511614

RESUMO

The comet assay or single cell gel electrophoresis has proven to be a versatile and sensitive method of measuring the induction and repair of DNA damage in individual cells. However, one of the drawbacks of the assay is the bias caused by changes in the ability of cells to repair DNA damage in different cell cycle phases. Whereas the bias seems less important when G0 peripheral blood lymphocytes are studied, it might cause problems when proliferating cells are investigated. In this paper, we validate the assumption that the total comet fluorescence intensity corresponds to the position of the cell in the cell cycle and can be used to assign single cells to specific cell cycle phases. To validate the approach, we used a very homogenous blood mononuclear CD34(+) cell population in G0 phase (unstimulated) or stimulated to enter the cell cycle. An analysis of the cell cycle distribution revealed that the 15 comet intensity classes and the 100 comets usually analyzed in a typical comet experiment are sufficient to obtain a reliable cell cycle distribution comparable with the results obtained by the flow cytometry for the same cell population. The effect of the cell cycle position on the results obtained by the comet assay for proliferating and non-proliferating cell populations irradiated with 3 Gy of X-radiation is also discussed.


Assuntos
Ciclo Celular , Ensaio Cometa/métodos , Dano ao DNA , Antígenos CD34/metabolismo , Ciclo Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Imunofenotipagem , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/efeitos da radiação , Raios X
11.
Cancers (Basel) ; 14(13)2022 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-35804898

RESUMO

In a living organism, cancer cells function in a specific microenvironment, where they exchange numerous physical and biochemical cues with other cells and the surrounding extracellular matrix (ECM). Immune evasion is a clinically relevant phenomenon, in which cancer cells are able to direct this interchange of signals against the immune effector cells and to generate an immunosuppressive environment favoring their own survival. A proper understanding of this phenomenon is substantial for generating more successful anticancer therapies. However, classical cell culture systems are unable to sufficiently recapture the dynamic nature and complexity of the tumor microenvironment (TME) to be of satisfactory use for comprehensive studies on mechanisms of tumor immune evasion. In turn, 3D-bioprinting is a rapidly evolving manufacture technique, in which it is possible to generate finely detailed structures comprised of multiple cell types and biomaterials serving as ECM-analogues. In this review, we focus on currently used 3D-bioprinting techniques, their applications in the TME research, and potential uses of 3D-bioprinting in modeling of tumor immune evasion and response to immunotherapies.

12.
Materials (Basel) ; 15(5)2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35268968

RESUMO

Poly-ɛ-caprolactone (PCL) is now widely studied in relation to the engineering of bone, cartilage, tendons, and other tissues. Standard histological protocols can destroy the carefully created trabecular and honeycomb-like architecture of PCL scaffolds, and could lead to scaffold fibers swelling, resulting in the displacement or compression of tissues inside the scaffold. The aim of this study was to modify a standard histopathological protocol for PCL scaffold preparation and evaluate it on porous cylindrical PCL scaffolds in a rat model. In 16 inbred Wag rats, 2 PCL scaffolds were implanted subcutaneously to both inguinal areas. Two months after implantation, harvested scaffolds were first subjected to µCT imaging, and then to histopathological analysis with standard (left inguinal area) and modified histopathological protocols (right inguinal area). To standardize the results, soft tissue percentages (STPs) were calculated on scaffold cross-sections obtained from both histopathological protocols and compared with corresponding µCT cross-sections. The modified protocol enabled the assessment of almost 10× more soft tissues on the scaffold cross-section than the standard procedure. Moreover, STP was only 1.5% lower than in the corresponding µCT cross-sections assessed before the histopathological procedure. The presented modification of the histopathological protocol is cheap, reproducible, and allows for a comprehensive evaluation of PCL scaffolds while maintaining their trabecular, honeycomb-like structure on cross-sections.

13.
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)
14.
Arch Med Sci ; 18(4): 1051-1061, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35832713

RESUMO

Introduction: Cytokines are important immune modulator factors controlling homeostasis of the body and are involved in tissue regeneration after wound healing. The encapsulation of cytokines in liposomes has many advantages potentially useful for their transfer to the cells. Liposomes protect cytokines from neutralization, improving their pharmacokinetics or biologic activity in vivo. They are targeted to specific cell types and may delay the release of cytokines, allowing their sustained paracrine delivery. Their physicochemical characteristics such as size, shape, charge, and stability are important parameters improving bio-distribution and prolonged pharmacokinetics of encapsulated cytokines. Material and methods: We developed an efficient protocol for the encapsulation of two types of cytokines, granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF), in liposomes that can be stored long term in the active state. Results: This method allows for the encapsulation of 12-13% of the total amount of cytokines and 50% of encapsulated cytokines are entrapped in liposomes of more than ≤ 600 nm in diameter. We show that in the studied cell lines the liposome-encapsulated cytokines do not affect cell morphology, proliferation or mortality. Conclusions: The G-CSF or GM-CSF can be delivered to the cells in working concentrations through the encapsulation in the liposomes. Before the clinical application, the efficiency of these liposomes should be confirmed by an in vivo study.

15.
Stem Cells Int ; 2021: 6665358, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093710

RESUMO

Clinical experiments suggest that mesenchymal stem cells (MSCs) may be useful for tissue repair therapies or treatment of the autoimmune disorders. There is still lack of consensus concerning the age limit of MSC donors, majority of researchers suggest the autologous MSC therapies of patients not exceeding age limit of 55-60 yrs. The purpose of our study was to compare the selected parameters of MSCs from adipose tissue (adipose stem cell, ASC) collected from young and old rats of ages corresponding to patient's ages 25 yrs. and 80 yrs., respectively. The differences of parameters of ASCs from young and old animals were compared with the differences between ASCs from short-term (3 passage) and long-term (30 passage) in vitro culture. Cell morphology, surface marker expression, growth potential, metabolic activity, ß-galactosidase activity, clonogenic potential, angiogenic potential, and differentiation ability of ASCs from young and aged animals and from in vitro cultures at 3rd and 30th passages were compared and analyzed. It may be concluded that ASCs may be applied for autologous transplantations in aged patients. Comparison of ASC aging dynamics depending on host aging or in vitro culture duration suggests that long-term in vitro culture may affect ASCs more than natural aging process of their host. We suggest that ASCs expanded in vitro prior to their clinical use must be carefully screened for the possible aging effects resulting not only from donor age, but from the duration of their in vitro culture.

16.
Rejuvenation Res ; 24(6): 441-448, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34918960

RESUMO

Mesenchymal stromal cells from adipose tissue (adipose stromal cells, ASCs) are regulators of repair processes in situ by paracrine mechanisms. These unique capabilities make ASCs candidates for the regenerative medicine applications, including cell-assisted lipotransfer method. ASC aging processes have been extensively researched in vitro, there is however limited information about the impact of ASC aging on their biological role in tissue regeneration in vivo. The aim of our study was the research of the possible effects of aging processes of ASCs resulting from the donor age or from in vitro aging during long-term culture (ASC expansion in bioreactors) on their capability to support survival of adipose subcutaneous transplants in rats. The supportive in vivo effects of ASCs from young donors were compared with the effects of ASCs from old donors and ASCs "aged" in long-term in vitro cultures. Fat grafts enriched with ASCs (regardless of their age) retain their volume longer than fat grafts without ASCs supplementation. Vascular expansion in cell-enriched fat grafts was more intense when compared with the controls. It may be concluded that the aging of ASCs does not substantially reduce their ability for the support of the survival of adipose tissue grafts.


Assuntos
Células-Tronco Mesenquimais , Tecido Adiposo , Animais , Técnicas de Cultura de Células , Ratos , Medicina Regenerativa
17.
Stem Cell Res Ther ; 11(1): 34, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31973733

RESUMO

BACKGROUND: Vascularization is important for the clinical application of tissue engineered products. Both adipose-derived stem cells (ASCs) and surgical prefabrication can be used to induce angiogenesis in scaffolds. Our aim was to compare the angiogenic potential of ASC-seeded scaffolds combined with scaffold prefabrication with that of non-seeded, non-prefabricated scaffolds. METHODS: For prefabrication, functional blood vessels were introduced into the scaffold using a flow-through pedicle system. ASCs were isolated from rat fat deposits. Three-dimensional-printed cylindrical poly-ε-caprolactone scaffolds were fabricated by fused deposition modelling. Three groups, each containing six rats, were investigated by using non-seeded, ASC-seeded, and osteogenic induced ASC-seeded scaffolds. In each group, one rat was implanted with two scaffolds in the inguinal region. On the right side, a scaffold was implanted subcutaneously around the inferior epigastric vessels (classic prefabrication group). On the left side, the inferior epigastric vessels were placed inside the prefabricated scaffold in the flow-through pedicle system (flow-through prefabrication group). The vessel density and vascular architecture were examined histopathologically and by µCT imaging, respectively, at 2 months after implantation. RESULTS: The mean vessel densities were 10- and 5-fold higher in the ASC-seeded and osteogenic induced ASC-seeded scaffolds with flow-through prefabrication, respectively, than in the non-seeded classic prefabricated group (p < 0.001). µCT imaging revealed functional vessels within the scaffold. CONCLUSION: ASC-seeded scaffolds with prefabrication showed significantly improved scaffold vasculogenesis and could be useful for application to tissue engineering products in the clinical settings.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Células-Tronco/metabolismo , Animais , Humanos , Neovascularização Fisiológica , Ratos , Alicerces Teciduais
18.
Cells ; 9(6)2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32560483

RESUMO

Under physiological conditions skeletal muscle regeneration depends on the satellite cells. After injury these cells become activated, proliferate, and differentiate into myofibers reconstructing damaged tissue. Under pathological conditions satellite cells are not sufficient to support regeneration. For this reason, other cells are sought to be used in cell therapies, and different factors are tested as a tool to improve the regenerative potential of such cells. Many studies are conducted using animal cells, omitting the necessity to learn about human cells and compare them to animal ones. Here, we analyze and compare the impact of IL-4 and SDF-1, factors chosen by us on the basis of their ability to support myogenic differentiation and cell migration, at mouse and human adipose tissue-derived stromal cells (ADSCs). Importantly, we documented that mouse and human ADSCs differ in certain reactions to IL-4 and SDF-1. In general, the selected factors impacted transcriptome of ADSCs and improved migration and fusion ability of cells in vitro. In vivo, after transplantation into injured muscles, mouse ADSCs more eagerly participated in new myofiber formation than the human ones. However, regardless of the origin, ADSCs alleviated immune response and supported muscle reconstruction, and cytokine treatment enhanced these effects. Thus, we documented that the presence of ADSCs improves skeletal muscle regeneration and this influence could be increased by cell pretreatment with IL-4 and SDF-1.


Assuntos
Quimiocina CXCL12/farmacologia , Interleucina-4/farmacologia , Mioblastos/citologia , Células Estromais/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Humanos , Camundongos , Regeneração/efeitos dos fármacos , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
19.
Acta Neurobiol Exp (Wars) ; 69(1): 46-51, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19325640

RESUMO

The finding of stem/progenitor cells in postnatal bone marrow and umbilical cord blood, opens up a possibility of using stem cells to treat neurologic diseases. There is a controversy, whether intravenously administered human umbilical cord blood cells (HUCBC) migrate to the brain, differentiate and improve recovery after ischemia. In this study, 1-3 x10;6 cells from non-cultured (non-committed) mononuclear HUCBC fraction were intravenously infused 1, 2, 3 or 7 days after a transient middle cerebral artery occlusion (MCAo) in adult rats. We found few human cells only in the ischemic area, localized mostly around blood vessels with few positive cells in the brain parenchyma. Timing of HUCBC delivery after ischemia or injection of Cyclosporin A at the time of delivery, had no effect on the number of human cells detected in the ischemic brain. Infusion of HUCBC did not reduce infarct volume and did not improve neurologic deficits after MCAo, suggesting that HUCBC failed to migrate/survive in the ischemic brain and did not provide significant neurological benefits.


Assuntos
Movimento Celular/fisiologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/citologia , Infarto da Artéria Cerebral Média/terapia , Células-Tronco/fisiologia , Análise de Variância , Animais , Biotinilação/métodos , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Humanos , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Infusões Intravenosas/métodos , Masculino , Exame Neurológico/métodos , Ratos , Ratos Wistar , Fatores de Tempo
20.
Stem Cells Int ; 2019: 1201927, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30915123

RESUMO

The purpose of the current study was to evaluate the usefulness of adipose-derived stem cells (ASCs) for bone injury therapy. Lipoaspirates were collected from the abdomen regions of 17 healthy female donors (mean age 49 ± 6 years) using Coleman technique or Body-jet liposuction. In the present study, the primary objective was the in vitro characteristics of human ASCs. The secondary objective was the optimization of the cell seeding process on 3D-printed scaffolds using polycaprolactone (PCL) or polycaprolactone covered with tricalcium phosphate (PCL + 5% TCP). Biological evaluation of human ASC showed high efficiency of isolation obtaining a satisfying amount of homogeneous cell populations. Results suggest that ASCs can be cultured in vitro for a long time without impairing their proliferative capacity. Growth kinetics shows that the highest number of cells can be achieved in passage 5 and after the 16th passage; there is a significant decrease of cell numbers and their proliferative potential. The percentage of colony forming units from the adipose stem cells is 8% ± 0.63% (p < 0.05). It was observed that the accumulation of calcium phosphate in the cells in vitro, marked with Alizarin Red S, was increased along with the next passage. Analysis of key parameters critically related to the cell seeding process shows that volume of cell suspension and propagation time greatly improve the efficiency of seeding both in PCL and PCL + 5% TCP scaffolds. The cell seeding efficiency did differ significantly between scaffold materials and cell seeding methods (p < 0.001). Increased seeding efficiency was observed when using the saturation of cell suspension into scaffolds with additional incubation. Alkaline phosphatase level production in PCL + 5% TCP scaffold was better than in PCL-only scaffold. The study results can be used for the optimization of the seeding process and quantification methods determining the successful implementation of the preclinical model study in the future tissue engineering strategies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA