Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
IUBMB Life ; 75(3): 196-206, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-34590780

RESUMO

The secretome of different cell types has been applied on in vitro and in vivo assays, indicating considerable therapeutic potential. However, the choice of the ideal cell type and culture conditions for obtaining the best set of soluble factors, as well as the assays to assess specific effects, remain subjects of vigorous debate. In this study, we used mass spectrometry to characterize the secretomes of ventricle derived-cardiac resident stromal cells (vCRSC) and human dermal fibroblasts (HDFs) and evaluate them in an effort to understand the niche specificity of biological responses toward different cellular behaviors, such as cell proliferation, adhesion, migration, and differentiation. It was interesting to note that the HDF and vCRSC secretomes were both able to induce proliferation and cardiac differentiation of H9c2 cells, as well as to increase the adhesion activity of H9c2 cells and human umbilical vein endothelial cells. Analysis of the secretome composition showed that the vCRSCs derived from different donors secreted a similar set of proteins. Despite the differences, almost half of the proteins identified in conditioned medium were common to both HDF and vCRSC. Consequently, a high number of common biological processes were identified in the secretomes of the two cell types, which could help to explain the similar results observed in the in vitro assays. We show that soluble factors secreted by both HDF and vCRSC are able to promote proliferation and differentiation of cardiomyoblasts in vitro. Our study indicates the possible use of vCRSC or HDF secretomes in acellular therapies for regenerative medicine.


Assuntos
Secretoma , Células Estromais , Humanos , Pele/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fibroblastos/metabolismo
2.
Wound Repair Regen ; 31(6): 827-841, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38038971

RESUMO

Therapies for wound healing using the secretome and extracellular vesicles (EVs) of mesenchymal stem/stromal cells have been shown to be successful in preclinical studies. This study aimed to characterise the protein content of the secretome from stem cells from human exfoliated deciduous teeth (SHED) and analyse the in vitro effects of SHED-conditioned medium (SHED-CM) and SHED extracellular vesicles (SHED-EVs) on keratinocytes. EVs were isolated and characterised. The keratinocyte viability and migration of cells treated with SHED-EVs and conditioned medium (CM) were evaluated. An HaCaT apoptosis model induced by H2 O2 in vitro was performed with H2 O2 followed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and live/dead assays. Finally, the expression of vascular endothelial growth factor (VEGF) in keratinocytes treated with secretome and EVs was evaluated by immunofluorescence staining and confirmed with RT-qPCR. SHED-EVs revealed a cup-shaped morphology with expression of the classical markers for exosomes CD9 and CD63, and a diameter of 181 ± 87 nm. The internalisation of EVs by HaCaT cells was confirmed by fluorescence microscopy. Proteomic analysis identified that SHED-CM is enriched with proteins related to stress response and development, including cytokines (CXCL8, IL-6, CSF1, CCL2) and growth factors (IGF2, MYDGF, PDGF). The results also indicated that 50% CM and 0.4-0.6 µg/mL EVs were similarly efficient for improving keratinocyte viability, migration, and attenuation of H2 O2 -induced cytotoxicity. Additionally, expression of VEGF on keratinocytes increased when treated with SHED secretome and EVs. Furthermore, VEGF gene expression in keratinocytes increased significantly when treated with SHED secretome and EVs. Both SHED-CM and SHED-EVs may therefore be promising therapeutic tools for accelerating re-epithelialization in wound healing.


Assuntos
Vesículas Extracelulares , Cicatrização , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Proteômica , Secretoma , Células-Tronco/metabolismo , Queratinócitos , Vesículas Extracelulares/metabolismo , Dente Decíduo
3.
Int J Mol Sci ; 23(11)2022 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-35682690

RESUMO

Cartilage repair has been a challenge in the medical field for many years. Although treatments that alleviate pain and injury are available, none can effectively regenerate the cartilage. Currently, regenerative medicine and tissue engineering are among the developed strategies to treat cartilage injury. The use of stem cells, associated or not with scaffolds, has shown potential in cartilage regeneration. However, it is currently known that the effect of stem cells occurs mainly through the secretion of paracrine factors that act on local cells. In this review, we will address the use of the secretome-a set of bioactive factors (soluble factors and extracellular vesicles) secreted by the cells-of mesenchymal stem cells as a treatment for cartilage regeneration. We will also discuss methodologies for priming the secretome to enhance the chondroregenerative potential. In addition, considering the difficulty of delivering therapies to the injured cartilage site, we will address works that use hydrogels functionalized with growth factors and secretome components. We aim to show that secretome-functionalized hydrogels can be an exciting approach to cell-free cartilage repair therapy.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Cartilagem/metabolismo , Cartilagem Articular/metabolismo , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/metabolismo , Secretoma , Engenharia Tecidual
4.
Int J Mol Sci ; 23(17)2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36077462

RESUMO

Endothelial-like cells may be obtained from CD133+ mononuclear cells isolated from human umbilical cord blood (hUCB) and expanded using endothelial-inducing medium (E-CD133 cells). Their use in regenerative medicine has been explored by the potential not only to form vessels but also by the secretion of bioactive elements. Extracellular vesicles (EVs) are prominent messengers of this paracrine activity, transporting bioactive molecules that may guide cellular response under different conditions. Using RNA-Seq, we characterized the miRNA content of EVs derived from E-CD133 cells cultivated under normoxia (N-EVs) and hypoxia (H-EVs) and observed that changing the O2 status led to variations in the selective loading of miRNAs in the EVs. In silico analysis showed that among the targets of differentially loaded miRNAs, there are transcripts involved in pathways related to cell growth and survival, such as FoxO and HIF-1 pathways. The data obtained reinforce the pro-regenerative potential of EVs obtained from E-CD133 cells and shows that fine tuning of their properties may be regulated by culture conditions.


Assuntos
Vesículas Extracelulares , MicroRNAs , Proliferação de Células , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Hipóxia/metabolismo , MicroRNAs/metabolismo
5.
BMC Genomics ; 20(1): 219, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30876407

RESUMO

BACKGROUND: Cardiac cell fate specification occurs through progressive steps, and its gene expression regulation features are still being defined. There has been an increasing interest in understanding the coordination between transcription and post-transcriptional regulation during the differentiation processes. Here, we took advantage of the polysome profiling technique to isolate and high-throughput sequence ribosome-free and polysome-bound RNAs during cardiomyogenesis. RESULTS: We showed that polysome-bound RNAs exhibit the cardiomyogenic commitment gene expression and that mesoderm-to-cardiac progenitor stages are strongly regulated. Additionally, we compared ribosome-free and polysome-bound RNAs and found that the post-transcriptional regulation vastly contributes to cardiac phenotype determination, including RNA recruitment to and dissociation from ribosomes. Moreover, we found that protein synthesis is decreased in cardiomyocytes compared to human embryonic stem-cells (hESCs), possibly due to the down-regulation of translation-related genes. CONCLUSIONS: Our data provided a powerful tool to investigate genes potentially controlled by post-transcriptional mechanisms during the cardiac differentiation of hESC. This work could prospect fundamental tools to develop new therapy and research approaches.


Assuntos
Biomarcadores/análise , Diferenciação Celular , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas/metabolismo , Miócitos Cardíacos/metabolismo , Polirribossomos/metabolismo , RNA Mensageiro/metabolismo , Células Cultivadas , Sequenciamento de Nucleotídeos em Larga Escala , Células-Tronco Embrionárias Humanas/citologia , Humanos , Miócitos Cardíacos/citologia , Organogênese , Polirribossomos/genética , RNA Mensageiro/genética
6.
Xenotransplantation ; 26(3): e12503, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30770594

RESUMO

Tissue-engineered heart valves aim to reproduce the biological properties of natural valves with anatomically correct structure and physiological performance. The closest alternative to creating an ideal heart valve substitute is to use decellularized porcine heart valves, due to their anatomy and availability. However, the immunological barrier and the structural maintenance limit the long-term physiological performance of decellularized porcine heart valves. This study investigated the extracellular matrix (ECM) structure of aortic and pulmonary porcine valves decellularized by a low concentration sodium dodecyl sulfate (SDS)-based method in order to determine the ECM scaffold (ECMS) conditions related to remodeling potential. To assess the structures of the leaflets and conduits of the heart valves, ECM components and their organization were evaluated by histology, biochemical analysis (BC), scanning electron microscopy, multiphoton microscopy, tensile test, immunofluorescence labeling (IF), and Raman microspectroscopy used to draw a profile of the cell niches. Histology and multiphoton imaging of decellularized aortic and pulmonary leaflets and conduits revealed a collagen and elastin histoarchitecture with rearrangement, loosening fibers, and glycosaminoglycan depletion confirmed by biochemistry quantification. The potential cytotoxicity of SDS residues was eliminated after 10 wash cycles. The mechanical properties of the structure of the valve indicated a functional resistance of decellularized ECM. The IF demonstrated the presence of basement membrane, suggesting a potential structure for host cell attachment. The RM analysis showed evidence of molecular interactions, suggesting conservation of the chemical composition, particularly among the protein molecular structures. The structural analyses performed in the semilunar porcine heart valves demonstrate that decellularized ECMS has structural properties that support physiological performance and potential host tissue integration. In fact, decellularized leaflet scaffolds were prone to cell interaction after human adipose-derived stromal cell seeding and culturing. Further analysis of biocompatibility, particularly the ECM-cell interaction, can elucidate the remodeling process, in preserved decellularized heart valve scaffold.


Assuntos
Próteses Valvulares Cardíacas , Valvas Cardíacas/cirurgia , Valva Pulmonar/cirurgia , Transplante Heterólogo , Animais , Valva Aórtica/cirurgia , Fenômenos Biomecânicos/fisiologia , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Feminino , Valvas Cardíacas/fisiologia , Humanos , Masculino , Suínos , Engenharia Tecidual/métodos
7.
Xenotransplantation ; 26(2): e12464, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30264494

RESUMO

Pericardial membrane derived from bovine heart tissues is a promising source of material for use in tissue-engineering applications. However, tissue processing is required for its use in humans due to the presence of animal antigens. Therefore, the purpose of this study was to evaluate the structural integrity and biocompatibility of the bovine pericardium (BP) after a soft decellularization process with a 0.1% sodium dodecyl sulfate (SDS) solution, with the aim to remove xenoantigens and preserve extracellular matrix (ECM) bioactivity. The decellularization process promoted a mean reduction of 77% of the amount of DNA in the samples in which cell nuclei staining was undetectable. The ECM content was maintained as mostly preserved after decellularization as well as its biomechanical properties. In addition, the decellularization protocol has proven to be efficient in removing the xenoantigen alpha-gal, which is responsible for immune rejection. The decellularized BP was noncytotoxic in vitro and allowed human adipose-derived stem cell (hASC) adhesion. Finally, after 7 days in culture, the tissue scaffold became repopulated by hASCs, and after 30 days, the ECM protein pro-collagen I was seen in the scaffold. Together, these characteristics indicated that soft BP decellularization with 0.1% SDS solution allows the acquirement of a bioactive scaffold suitable for cell repopulation and potentially useful for regenerative medicine.


Assuntos
Matriz Extracelular/imunologia , Pericárdio/imunologia , Engenharia Tecidual , Alicerces Teciduais , Animais , Bovinos , Matriz Extracelular/metabolismo , Humanos , Dodecilsulfato de Sódio/metabolismo , Engenharia Tecidual/métodos , Transplante Heterólogo/métodos
8.
Int J Mol Sci ; 20(6)2019 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-30875722

RESUMO

Extracellular vesicles (EVs) are particles released from different cell types and represent key components of paracrine secretion. Accumulating evidence supports the beneficial effects of EVs for tissue regeneration. In this study, discarded human heart tissues were used to isolate human heart-derived extracellular vesicles (hH-EVs). We used nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) to physically characterize hH-EVs and mass spectrometry (MS) to profile the protein content in these particles. The MS analysis identified a total of 1248 proteins. Gene ontology (GO) enrichment analysis in hH-EVs revealed the proteins involved in processes, such as the regulation of cell death and response to wounding. The potential of hH-EVs to induce proliferation, adhesion, angiogenesis and wound healing was investigated in vitro. Our findings demonstrate that hH-EVs have the potential to induce proliferation and angiogenesis in endothelial cells, improve wound healing and reduce mesenchymal stem-cell adhesion. Last, we showed that hH-EVs were able to significantly promote mesenchymal stem-cell recellularization of decellularized porcine heart valve leaflets. Altogether our data confirmed that hH-EVs modulate cellular processes, shedding light on the potential of these particles for tissue regeneration and for scaffold recellularization.


Assuntos
Vesículas Extracelulares/metabolismo , Valvas Cardíacas/química , Células-Tronco Mesenquimais/citologia , Proteínas/metabolismo , Proteômica/métodos , Animais , Brasil , Adesão Celular , Proliferação de Células , Células Cultivadas , Ontologia Genética , Valvas Cardíacas/metabolismo , Humanos , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Nanotecnologia , Neovascularização Fisiológica , Suínos , Bancos de Tecidos
9.
Cells Tissues Organs ; 203(3): 173-182, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27802444

RESUMO

The tissue microenvironment regulates such stem cell behaviors as self-renewal and differentiation. Attempts to mimic components of these microenvironments could provide new strategies for culturing and directing the behavior of stem cells. The aim of the present study was to mimic cardiac and umbilical cord tissue microenvironments in vitro and compare the resulting bone marrow-derived mesenchymal stem cell (BM-MSC) behaviors. We generated tissue microenvironments using conditioned medium (CM) and extracellular matrix (ECM) samples obtained from human heart and umbilical cord tissue explant cultures and by tissue decellularization. Mass spectrometry and immunostaining were used to characterize and determine the specific protein profiles of the ECMs and CMs. We demonstrated that the ECMs and CMs were not cytotoxic to BM-MSCs and could thus be tested via cell culture. The BM-MSCs showed a higher proliferation rate when cultured with umbilical cord-derived CM compared with the other analyzed conditions. Furthermore, the ECMs increased cell adhesion and migration. However, although the conditions tested in this work were able to maintain the viability and affect the proliferation, adhesion and migration of BM-MSCs in vitro, mimicking tissue microenvironments using ECM and CM was not sufficient to induce the cardiomyogenic differentiation of BM-MSCs. The present study provides a thorough characterization of the biological activity of these ECMs and CMs in human BM-MSC cultures.


Assuntos
Microambiente Celular , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Transdução de Sinais , Cordão Umbilical/citologia , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
BMC Mol Biol ; 15: 12, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24993635

RESUMO

BACKGROUND: DZIP1 (DAZ-interacting protein 1) has been described as a component of the Hh signaling pathway with a putative regulatory role in ciliogenesis. DZIP1 interacts with DAZ RNA binding proteins in embryonic stem cells and human germ cells suggesting a role in mRNA regulation. RESULTS: We investigated DZIP1 function in HeLa cells and its involvement in ribonucleoprotein complexes. DZIP1 was predominantly located in granules in the cytoplasm. Under oxidative stress conditions, DZIP1 re-localized to stress granules. DZIP appears to be important for the formation of stress granules during the stress response. We used immunoprecipitation assays with antibodies against DZIP1 and microarray hybridization to identify mRNAs associated with DZIP1. The genetic networks formed by the DZIP1-associated mRNAs were involved in cell cycle and gene expression regulation. DZIP1 is involved in the Hedgehog signaling pathway. We used cyclopamine, a specific inhibitor of this pathway, to analyze the expression of DZIP1 and its associated mRNAs. The abundance of DZIP1-associated mRNAs increased with treatment; however, the silencing or overexpression of DZIP1 in HeLa cells had no effect on the accumulation of the associated mRNAs. Polysomal profile analysis by sucrose gradient centrifugation demonstrated the presence of DZIP1 in the polysomal fraction. CONCLUSIONS: Our results suggest that DZIP1 is part of an RNP complex that occupies various subcellular locations. The diversity of the mRNAs associated with DZIP1 suggests that this protein is a component of different RNPs associated with translating polysomes and with RNA granules.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Grânulos Citoplasmáticos/genética , Estresse Oxidativo/genética , Ribonucleoproteínas/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Regulação da Expressão Gênica/genética , Células HeLa , Proteínas Hedgehog/genética , Humanos , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais/genética
11.
Stem Cell Res Ther ; 15(1): 15, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38229157

RESUMO

BACKGROUND: Although the paracrine effects of mesenchymal stem/stromal cells (MSCs) have been recognized as crucial mediators of their regenerative effects on tissue repair, the potential of MSC secretomes as effective substitutes for cellular therapies remains underexplored. METHODS: In this study, we compared MSCs from the human dermis (DSCs) and adipose tissue (ASCs) with their secretomes regarding their efficacy for skin wound healing using a translationally relevant murine model. RESULTS: Proteomic analysis revealed that while there was a substantial overlap in protein composition between DSC and ASC secretomes, specific proteins associated with wound healing and angiogenesis were differentially expressed. Despite a similar angiogenic potential in vivo, DSC and ASC secretomes were found to be less effective than cells in accelerating wound closure and promoting tissue remodeling. CONCLUSIONS: Overall, secretome-treated groups showed intermediary results between cells- and control-treated (empty scaffold) groups. These findings highlight that although secretomes possess therapeutic potential, their efficacy might be limited compared to cellular therapies. This study contributes to the growing understanding of MSC secretomes, emphasizes the need for further protocol optimization, and offers insights into their potential applications in regenerative medicine.


Assuntos
Células-Tronco Mesenquimais , Secretoma , Humanos , Animais , Camundongos , Proteômica/métodos , Tecido Adiposo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Cicatrização
12.
FASEB J ; 26(11): 4390-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22815386

RESUMO

Cell migration is a crucial event for normal T-cell development, and various ligand/receptor pairs have been implicated. Most of them, including chemokines and extracellular matrix proteins, have attractant properties on thymocytes. We discuss herein two further groups of ligand/receptor pairs, semaphorins/neuropilins and ephs/ephrins, which are constitutively expressed by thymocytes and thymic microenvironmental cells. Evidence shows that the corresponding interactions are relevant for developing T-cell migration, including the entry of bone marrow progenitor cells, migration of CD4/CD8-defined thymocyte subpopulations triggered by chemokines and/or extracellular matrix proteins, and thymocyte export. Conceptually, the data summarized here show that thymocyte migration results from a complex network of molecular interactions, which generate not only attraction, but also repulsion of migrating T-cell precursors.


Assuntos
Movimento Celular/fisiologia , Efrinas/metabolismo , Semaforinas/metabolismo , Timócitos/fisiologia , Animais , Efrinas/genética , Regulação da Expressão Gênica , Semaforinas/genética
13.
Front Bioeng Biotechnol ; 11: 1304030, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38260748

RESUMO

Articular cartilage is a highly specialized tissue able to tolerate physical stress. However, its capacity for restoration is restricted, and injuries to the cartilage do not recover spontaneously. Interest in mesenchymal stem cells derived from human adipose tissue (hASCs) is growing due to their potential to improve tissue healing and recovery. Decellularized extracellular matrix (dECM)-based hydrogels combined with hASCs could serve as an interface for studying behavior and differentiation properties in a cartilage microenvironment. In the present study, we described the behavior of hASCs cultured in a commercial dECM MatriXpec™. The structural microtopography of MatriXpec™ was analyzed by scanning electron micrography, and its protein composition was accessed by mass spectrometry. The protein composition of MatriXpec™ is mainly represented by collagen proteins, building its fibrous ultrastructure. hASCs were cultured three-dimensionally (3D) on MatriXpec™ to perform cell viability, growth, and cartilage differentiation analysis. We showed that MatriXpec™ could be loaded with hASCs and that it supports cell maintenance for several days. We observed that the three-dimensional ultrastructure of the biomaterial is composed of nanofibers, and its protein composition reflects the tissue from which it was harvested. Finally, we showed that the molecular cues from the hydrogel are biologically active as these influence cell behavior and differentiation phenotype, increasing the expression of fibrocartilage-related genes such as SOX9, COL1, COL10, and MMP13. MatriXpec™ hydrogel can be used as an interface for 3D hASCs culture studies as it maintains cell viability and supports its differentiation process.

14.
World J Stem Cells ; 15(5): 453-465, 2023 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-37342215

RESUMO

Extracellular vesicles (EVs) are nanometric particles that enclose cell-derived bioactive molecules in a lipid bilayer and serve as intercellular communication tools. Accordingly, in various biological contexts, EVs are reported to engage in immune modulation, senescence, and cell proliferation and differentiation. Therefore, EVs could be key elements for potential off-the-shelf cell-free therapy. Little has been studied regarding EVs derived from human pluripotent stem cells (hPSC-EVs), even though hPSCs offer good opportunities for induction of tissue regeneration and unlimited proliferative ability. In this review article, we provide an overview of studies using hPSC-EVs, focusing on identifying the conditions in which the cells are cultivated for the isolation of EVs, how they are characterized, and applications already demonstrated. The topics reported in this article highlight the incipient status of the studies in the field and the significance of hPSC-EVs' prospective applications as PSC-derived cell-free therapy products.

15.
Polymers (Basel) ; 15(24)2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38139882

RESUMO

Articular cartilage injuries are inherently irreversible, even with the advancement in current therapeutic options. Alternative approaches, such as the use of mesenchymal stem/stromal cells (MSCs) and tissue engineering techniques, have gained prominence. MSCs represent an ideal source of cells due to their low immunogenicity, paracrine activity, and ability to differentiate. Among biomaterials, self-assembling peptide hydrogels (SAPH) are interesting given their characteristics such as good biocompatibility and tunable properties. Herein we associate human adipose-derived stem cells (hASCs) with a commercial SAPH, Puramatrix™, to evaluate how this three-dimensional microenvironment affects cell behavior and its ability to undergo chondrogenic differentiation. We demonstrate that the Puramatrix™ hydrogel comprises a highly porous matrix permissible for hASC adhesion and in vitro expansion. The morphology and cell growth dynamics of hASCs were affected when cultured on the hydrogel but had minimal alteration in their immunophenotype. Interestingly, hASCs spontaneously formed cell aggregates throughout culturing. Analysis of glycosaminoglycan production and gene expression revealed a noteworthy and donor-dependent trend suggesting that Puramatrix™ hydrogel may have a natural capacity to support the chondrogenic differentiation of hASCs. Altogether, the results provide a more comprehensive understanding of the potential applications and limitations of the Puramatrix™ hydrogel in developing functional cartilage tissue constructs.

16.
Front Bioeng Biotechnol ; 11: 1297327, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38239914

RESUMO

Bone lesions affect individuals of different age groups, compromising their daily activities and potentially leading to prolonged morbidity. Over the years, new compositions and manufacturing technologies were developed to offer customized solutions to replace injured tissue and stimulate tissue regeneration. This work used digital light processing (DPL) technology for three-dimensional (3D) printing of porous structures using pre-ceramic polymer, followed by pyrolysis to obtain SiOC vitreous scaffolds. The SiOC scaffolds produced had an amorphous structure (compatible with glass) with an average porosity of 72.69% ± 0.99, an average hardness of 935.1 ± 71.0 HV, and an average maximum flexural stress of 7.8 ± 1.0 MPa, similar to cancellous bone tissue. The scaffolds were not cytotoxic and allowed adult stem cell adhesion, growth, and expansion. After treatment with osteoinductive medium, adult stem cells in the SiOC scaffolds differentiated to osteoblasts, assuming a tissue-like structure, with organization in multiple layers and production of a dense fibrous matrix rich in hydroxyapatite. The in vitro analyses supported the hypothesis that the SiOC scaffolds produced in this work were suitable for use as a bone substitute for treating critically sized lesions, with the potential to stimulate the gradual process of regeneration of the native tissue. The data obtained stimulate the continuity of studies with the SiOC scaffolds developed in this work, paving the way for evaluating safety and biological activity in vivo.

17.
Stem Cell Rev Rep ; 18(1): 94-125, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34545529

RESUMO

Mesenchymal stem/stromal cells (MSCs) are remarkable tools for regenerative medicine. Therapeutic approaches using these cells can promote increased activity and viability in several cell types through diverse mechanisms such as paracrine and immunomodulatory activities, contributing substantially to tissue regeneration and functional recovery. However, biological samples of human MSCs, usually obtained from adult tissues, often exhibit variable behavior during in vitro culture, especially with respect to cell population heterogeneity, replicative senescence, and consequent loss of functionality. Accordingly, it is necessary to establish standard protocols to generate high-quality, stable cell cultures, for example, by using pluripotent stem cells (PSCs) in derivation protocols of MSC-like cells since PSCs maintain their characteristics consistently during culture. However, the available protocols seem to generate distinct populations of PSC-derivedMSCs (PSC-MSCs) with peculiar attributes, which do not always resemble bona fide primary MSCs. The present review addresses the developmental basis behind some of these derivation protocols, exposing the differences among them and discussing the functional properties of PSC-MSCs, shedding light on elements that may help determine standard characterizations and criteria to evaluate and define these cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Células-Tronco Pluripotentes , Diferenciação Celular , Humanos , Medicina Regenerativa/métodos
18.
Data Brief ; 41: 107963, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35242943

RESUMO

The study of the secretome of different cell types has gained prominence over the years due to its role in understanding the cell microenvironment and possible uses in acellular therapies. Approaches in this field include proteomic characterizations of the secretomes as well as evaluating their potential to induce cell and tissue responses. Here, we present the mass spectrometry proteomics data from a characterization of the secretome of cardiac resident stromal cells (CRSCs) and dermal fibroblasts in order to compare their compositions. To evaluate the potential for cell proliferation, differentiation, migration, and adhesion, in vitro assays were performed and analyzed using a high-content imaging system. For each assay, specific analysis strategies were developed to quantify the generated data. These datasets provide insights into the differences and similarities between secretomes from different cell sources. It also describes methodologies for analyzing images from different in vitro assays using high-throughput automated imaging.

19.
Stem Cells Int ; 2022: 4930932, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047041

RESUMO

Dysfunctions in adipose tissue cells are responsible for several obesity-related metabolic diseases. Understanding the process of adipocyte formation is thus fundamental for understanding these diseases. The adipocyte differentiation of adipose-derived stem/stromal cells (ADSCs) showed a reduction in the mRNA level of the interleukin 21 receptor (IL21R) during this process. Although the receptor has been associated with metabolic diseases, few studies have examined its function in stem cells. In this study, we used confocal immunofluorescence assays to determine that IL21R colocalizes with mitochondrial protein ATP5B, ALDH4A1, and the nucleus of human ADSCs. We demonstrated that silencing and overexpression of IL21R did not affect the cell proliferation and mitochondrial activity of ADSCs. However, IL21R silencing did reduce ADSC adipogenic capacity. Further studies are needed to understand the mechanism involved between IL21R and the adipogenic differentiation process.

20.
Stem Cell Res Ther ; 13(1): 122, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35313959

RESUMO

BACKGROUND: COVID-19 is a multisystem disease that presents acute and persistent symptoms, the postacute sequelae (PASC). Long-term symptoms may be due to consequences from organ or tissue injury caused by SARS-CoV-2, associated clotting or inflammatory processes during acute COVID-19. Various strategies are being chosen by clinicians to prevent severe cases of COVID-19; however, a single treatment would not be efficient in treating such a complex disease. Mesenchymal stromal cells (MSCs) are known for their immunomodulatory properties and regeneration ability; therefore, they are a promising tool for treating disorders involving immune dysregulation and extensive tissue damage, as is the case with COVID-19. This study aimed to assess the safety and explore the long-term efficacy of three intravenous doses of UC-MSCs (umbilical cord MSCs) as an adjunctive therapy in the recovery and postacute sequelae reduction caused by COVID-19. To our knowledge, this is one of the few reports that presents the longest follow-up after MSC treatment in COVID-19 patients. METHODS: This was a phase I/II, prospective, single-center, randomized, double-blind, placebo-controlled clinical trial. Seventeen patients diagnosed with COVID-19 who require intensive care surveillance and invasive mechanical ventilation-critically ill patients-were included. The patient infusion was three doses of 5 × 105 cells/kg UC-MSCs, with a dosing interval of 48 h (n = 11) or placebo (n = 6). The evaluations consisted of a clinical assessment, viral load, laboratory testing, including blood count, serologic, biochemical, cell subpopulation, cytokines and CT scan. RESULTS: The results revealed that in the UC-MSC group, there was a reduction in the levels of ferritin, IL-6 and MCP1-CCL2 on the fourteen day. In the second month, a decrease in the levels of reactive C-protein, D-dimer and neutrophils and an increase in the numbers of TCD3, TCD4 and NK lymphocytes were observed. A decrease in extension of lung damage was observed at the fourth month. The improvement in all these parameters was maintained until the end of patient follow-up. CONCLUSIONS: UC-MSCs infusion is safe and can play an important role as an adjunctive therapy, both in the early stages, preventing severe complications and in the chronic phase with postacute sequelae reduction in critically ill COVID-19 patients. Trial registration Brazilian Registry of Clinical Trials (ReBEC), UTN code-U1111-1254-9819. Registered 31 October 2020-Retrospectively registered, https://ensaiosclinicos.gov.br/rg/RBR-3fz9yr.


Assuntos
COVID-19 , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Estudos Prospectivos , SARS-CoV-2
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa