Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Mol Med ; 22(6): 2964-2969, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29536627

RESUMO

Biofabrication of tissue analogues is aspiring to become a disruptive technology capable to solve standing biomedical problems, from generation of improved tissue models for drug testing to alleviation of the shortage of organs for transplantation. Arguably, the most powerful tool of this revolution is bioprinting, understood as the assembling of cells with biomaterials in three-dimensional structures. It is less appreciated, however, that bioprinting is not a uniform methodology, but comprises a variety of approaches. These can be broadly classified in two categories, based on the use or not of supporting biomaterials (known as "scaffolds," usually printable hydrogels also called "bioinks"). Importantly, several limitations of scaffold-dependent bioprinting can be avoided by the "scaffold-free" methods. In this overview, we comparatively present these approaches and highlight the rapidly evolving scaffold-free bioprinting, as applied to cardiovascular tissue engineering.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Bioimpressão/tendências , Doenças Cardiovasculares/terapia , Sistema Cardiovascular/fisiopatologia , Doenças Cardiovasculares/fisiopatologia , Humanos , Hidrogéis/uso terapêutico , Engenharia Tecidual/tendências , Alicerces Teciduais
2.
Stem Cells ; 35(5): 1273-1289, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28233376

RESUMO

Adipose tissue is a rich source of multipotent mesenchymal stem-like cells, located in the perivascular niche. Based on their surface markers, these have been assigned to two main categories: CD31- /CD45- /CD34+ /CD146- cells (adventitial stromal/stem cells [ASCs]) and CD31- /CD45- /CD34- /CD146+ cells (pericytes [PCs]). These populations display heterogeneity of unknown significance. We hypothesized that aldehyde dehydrogenase (ALDH) activity, a functional marker of primitivity, could help to better define ASC and PC subclasses. To this end, the stromal vascular fraction from a human lipoaspirate was simultaneously stained with fluorescent antibodies to CD31, CD45, CD34, and CD146 antigens and the ALDH substrate Aldefluor, then sorted by fluorescence-activated cell sorting. Individual ASCs (n = 67) and PCs (n = 73) selected from the extremities of the ALDH-staining spectrum were transcriptionally profiled by Fluidigm single-cell quantitative polymerase chain reaction for a predefined set (n = 429) of marker genes. To these single-cell data, we applied differential expression and principal component and clustering analysis, as well as an original gene coexpression network reconstruction algorithm. Despite the stochasticity at the single-cell level, covariation of gene expression analysis yielded multiple network connectivity parameters suggesting that these perivascular progenitor cell subclasses possess the following order of maturity: (a) ALDHbr ASC (most primitive); (b) ALDHdim ASC; (c) ALDHbr PC; (d) ALDHdim PC (least primitive). This order was independently supported by specific combinations of class-specific expressed genes and further confirmed by the analysis of associated signaling pathways. In conclusion, single-cell transcriptional analysis of four populations isolated from fat by surface markers and enzyme activity suggests a developmental hierarchy among perivascular mesenchymal stem cells supported by markers and coexpression networks. Stem Cells 2017;35:1273-1289.


Assuntos
Tecido Adiposo/citologia , Linhagem da Célula , Redes Reguladoras de Genes , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Aldeído Desidrogenase/metabolismo , Diferenciação Celular/genética , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Pericitos/citologia , Análise de Célula Única
3.
Biotechnol Lett ; 40(8): 1189-1200, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29876793

RESUMO

OBJECTIVES: Attachment of magnetic particles to cells is needed for a variety of applications but is not always possible or efficient. Simpler and more convenient methods are thus desirable. In this study, we tested the hypothesis that endothelial cells (EC) can be loaded with micron-size magnetic beads by the phagocytosis-like mechanism 'angiophagy'. To this end, human umbilical vein EC (HUVEC) were incubated with magnetic beads conjugated or not (control) with an anti-VEGF receptor 2 antibody, either in suspension, or in culture followed by re-suspension using trypsinization. RESULTS: In all conditions tested, HUVEC incubation with beads induced their uptake by angiophagy, which was confirmed by (i) increased cell granularity assessed by flow cytometry, and (ii) the presence of an F-actin rich layer around many of the intracellular beads, visualized by confocal microscopy. For confluent cultures, the average number of beads per cell was 4.4 and 4.2, with and without the presence of the anti-VEGFR2 antibody, respectively. However, while the actively dividing cells took up 2.9 unconjugated beads on average, this number increased to 5.2 if binding was mediated by the antibody. Magnetic pulldown increased the cell density of beads-loaded cells in porous electrospun poly-capro-lactone scaffolds by a factor of 4.5 after 5 min, as compared to gravitational settling (p < 0.0001). CONCLUSION: We demonstrated that EC can be readily loaded by angiophagy with micron-sized beads while attached in monolayer culture, then dispersed in single-cell suspensions for pulldown in porous scaffolds and for other applications.


Assuntos
Endocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Magnetismo , Microesferas , Coloração e Rotulagem/métodos , Citometria de Fluxo , Humanos , Microscopia Confocal
4.
Tissue Eng Part C Methods ; 27(3): 225-231, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33446076

RESUMO

Three-dimensional (3D) bioprinting is an emerging tissue engineering technology, already with several remarkable accomplishments and with more promises to fulfill. Besides the enduring goal of making tissues for implantation, it could also become an essential tool in the worldwide trend to replace animal experimentation with improved in vitro models for disease mechanism studies, or with new high-throughput pharmacological and toxicology assays. All these require the speed, reproducibility, and standardization that bioprinting could easily provide. However, originating from additive manufacturing with its top-down approach of "filling" a virtual volume with a semifluid (hydrogel) material, the finer internal anatomic structure of the tissues, as well as vascularization and innervation, has remained difficult to implement. Thus, the next frontier in bioprinting is the generation of more anatomically realistic models, needed for ascending to the functionality of living tissues. In this study, I discuss the conceptual and practical barriers still hampering the attainment of this goal and suggest solutions to overcome them. In this regard, I introduce two workflows that combine existing methods in new operational sequences: (1) bioprinting guided by images of histological sections assembled in 3D constructs and (2) bioprinting of bidimensional vascular patterns implemented among stackable cellular layers. While more sophisticated methods to capture the tissue structure in 3D constructs certainly exist, I contend that extrusion bioprinting may still offer a simple, practical, and affordable option. Impact statement Paucity of anatomic structural details is one of the limitations of three-dimensional bioprinting toward fulfilling its potential for tissue engineering, drug testing, and toxicological assays. The origins of this problem can be tracked back to derivation of bioprinting from inorganic additive manufacturing, making it more adept to render the shapes of the objects than their content. As solutions, I suggest two simple workflows that can be implemented by most current bioprinters, based on the import into the construct design of anatomically realistic structural information. If more largely adopted, these and similar approaches may significantly improve the applicability of bioprinted constructs.


Assuntos
Bioimpressão , Animais , Hidrogéis , Impressão Tridimensional , Reprodutibilidade dos Testes , Engenharia Tecidual , Alicerces Teciduais
5.
Biofabrication ; 13(3)2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33498017

RESUMO

In vitrotumor models consisting of cell spheroids are increasingly used for mechanistic studies and pharmacological testing. However, unless vascularized, the availability of nutrients such as glucose to deeper layers of multicellular aggregates is limited. In addition, recent developments in cells-only biofabrication (e.g. 'scaffold-free bioprinting'), allow the creation of more complex spheroid-based structures, further exposing the cells to nutrient deprivation within these constructs. To explore the impact of glucose availability on such tumor-like structures, we used the CompuCell3D platform for modeling of tumor spheroids. By monitoring the types of cells, fusing pairs geometry and the distance between spheroids centers of mass, we made novel heuristic observations on how binary- and multi-spheroid fusions are impacted by glucose availability. At limiting glucose concentrations mimicking hypoglycemia we noted an abrupt collapse of the tumor spheroids, unexpectedly amplified by the contact with normal cell spheroids. At higher glucose concentrations, we found an increased intermixing of cancerous cells, strong anti-phase oscillations between proliferating and quiescent tumor cells and a structural instability of fusing tumor spheroids, leading to their re-fragmentation. In a model of tumor microenvironment composed of normal cell spheroids fusing around a tumoral one, the competition for glucose lead to either the tumor's disappearance, to a steady state, or to its expansion. Moreover, the invasion of this microenvironment by individual tumor cells was also strongly depended on the available glucose. In conclusion, we demonstrate the value of computational simulations for anticipating the properties of biofabricated tumor models, and in generating testable hypotheses regarding the relationship between cancer, nutrition and diabetes.


Assuntos
Bioimpressão , Neoplasias , Simulação por Computador , Glucose , Humanos , Esferoides Celulares , Microambiente Tumoral
6.
Am J Physiol Heart Circ Physiol ; 298(6): H1832-41, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20363884

RESUMO

Treating cancer patients with chemotherapeutics, such as doxorubicin (Dox), cause dilated cardiomyopathy and congestive heart failure because of oxidative stress. On the other hand, heat shock factor-1 (HSF-1), a transcription factor for heat shock proteins (Hsps), is also known to be activated in response to oxidative stress. However, the possible role of HSF-1 activation and the resultant Hsp25 in chemotherapeutic-induced heart failure has not been investigated. Using HSF-1 wild-type (HSF-1(+/+)) and knock-out (HSF-1(-/-)) mice, we tested the hypothesis that activation of HSF-1 plays a role in the development of Dox-induced heart failure. Higher levels of Hsp25 and its phosphorylated forms were found in the failing hearts of Dox-treated HSF-1(+/+) mice. More than twofold increase in Hsp25 mRNA level was found in Dox-treated hearts. Proteomic analysis showed that there is accumulation and aggregation of Hsp25 in Dox-treated failing hearts. Additionally, Hsp25 was found to coimmunoprecipitate with p53 and vice versa. Further studies indicated that the Dox-induced higher levels of Hsp25 transactivated p53 leading to higher levels of the pro-apoptotic protein Bax, but other p53-related proteins remained unaltered. Moreover, HSF-1(-/-) mice showed significantly reduced Dox-induced heart failure and higher survival rate, and there was no change in Bax upon treating with Dox in HSF-1(-/-) mice. From these results we propose a novel mechanism for Dox-induced heart failure: increased expression of Hsp25 because of oxidant-induced activation of HSF-1 transactivates p53 to increase Bax levels, which leads to heart failure.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Proteínas de Ligação a DNA/fisiologia , Doxorrubicina/efeitos adversos , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/fisiopatologia , Fatores de Transcrição/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Insuficiência Cardíaca/metabolismo , Fatores de Transcrição de Choque Térmico , Proteínas de Choque Térmico/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo/fisiologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
7.
Microvasc Res ; 79(3): 207-16, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20188747

RESUMO

Bone-marrow-derived endothelial progenitor cells (EPCs) contribute to angiogenesis-mediated pathological neovascularization, and recent studies have begun to recognize the biological significance of this contribution. This review will discuss the ability of EPCs to contribute to neovascularization in both physiological and pathological conditions. Circulating EPCs were originally identified in 1997 by Asahara as CD34(+) VEGFR2(+) mononuclear cells. These cells differentiated into an endothelial phenotype, expressed endothelial markers, and incorporated into neovessels at sites of ischemia (Asahara et al., 1997). EPCs provide both instructive (release of pro-angiogenic cytokines) and structural (vessel incorporation and stabilization) functions that contribute to the initiation of neo-angiogenesis. EPC populations can be characterized based on surface markers of freshly isolated cells, or they can be described by their in vitro characteristics once placed in culture. However, a major stumbling block to progress in the field has been the lack of consensus among investigators as to the optimal characterization of EPCs. This review intends to address the role of both EPC classes and evaluate how they interact in the setting of pathological angiogenesis. Since the EPCs may be responsible for turning on the "angiogenic switch," strategies have been employed to keep this switch in the "off" position for diseases like cancer, retinopathy, and wet AMD. The expectation is that EPCs will evolve into clinically useful prognostic and predictive tools in cancer and in ocular diseases associated with pathological neovascularization and that targeting this cell type is a key to successful management of patients suffering from diseases associated with pathological neovascularization.


Assuntos
Células Endoteliais/patologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica/patologia , Células-Tronco/patologia , Inibidores da Angiogênese/uso terapêutico , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Endoteliais/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica , Fenótipo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/fisiopatologia , Nicho de Células-Tronco , Células-Tronco/efeitos dos fármacos
8.
Mol Ther ; 17(9): 1594-604, 2009 09.
Artigo em Inglês | MEDLINE | ID: mdl-19584817

RESUMO

The ability to control the differentiation of adult hematopoietic stem cells (HSCs) would promote development of new cell-based therapies to treat multiple degenerative diseases. Systemic injection of NaIO(3) was used to ablate the retinal pigment epithelial (RPE) layer in C57Bl6 mice and initiate neural retinal degeneration. HSCs infected ex vivo with lentiviral vector expressing the RPE-specific gene RPE65 restored a functional RPE layer, with typical RPE phenotype including coexpression of another RPE-specific marker, CRALBP, and photoreceptor outer segment phagocytosis. Retinal degeneration was prevented and visual function, as measured by electroretinography (ERG), was restored to levels similar to that found in normal animals. None of the controls (no HSCs, HSCs alone and HSCs infected with lentiviral vector expressing LacZ) showed these effects. In vitro gene array studies demonstrated that infection of HSC with RPE65 increased adenylate cyclase mRNA. In vitro exposure of HSCs to a pharmacological agonist of adenylate cyclase also led to in vitro differentiation of HSCs to RPE-like cells expressing pigment granules and the RPE-specific marker, CRALBP. Our data confirm that expression of the cell-specific gene RPE65 promoted fate determination of HSCs toward RPE for targeted tissue repair, and did so in part by activation of adenylate cyclase signaling pathways. Expression by HSCs of single genes unique to a differentiated cell may represent a novel experimental paradigm to influence HSC plasticity, force selective differentiation, and ultimately lead to identification of pharmacological alternatives to viral gene delivery.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Diferenciação Celular , Células Cultivadas , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Feminino , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imuno-Histoquímica , Lentivirus/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/terapia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/ultraestrutura , cis-trans-Isomerases
9.
Int J Bioprint ; 5(1): 167, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32596531

RESUMO

The overarching principle of three-dimensional (3D) bioprinting is the placing of cells or cell clusters in the 3D space to generate a cohesive tissue microarchitecture that comes close to in vivo characteristics. To achieve this goal, several technical solutions are available, generating considerable combinatorial bandwidth: (i) Support structures are generated first, and cells are seeded subsequently; (ii) alternatively, cells are delivered in a printing medium, so-called "bioink," that contains them during the printing process and ensures shape fidelity of the generated structure; and (iii) a "scaffold-free" version of bioprinting, where only cells are used and the extracellular matrix is produced by the cells themselves, also recently entered a phase of accelerated development and successful applications. However, the scaffold-free approaches may still benefit from secondary incorporation of scaffolding materials, thus expanding their versatility. Reversibly, the bioink-based bioprinting could also be improved by adopting some of the principles and practices of scaffold-free biofabrication. Collectively, we anticipate that combinations of these complementary methods in a "hybrid" approach, rather than their development in separate technological niches, will largely increase their efficiency and applicability in tissue engineering.

10.
Mol Vis ; 14: 1064-70, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18552982

RESUMO

PURPOSE: It is crucial for the treatment of severe ocular surface diseases such as Stevens-Johnson syndrome (SJS) and ocular cicatricial pemphigoid (OCP) to find strategies that avoid the risks of allograft rejection and immunosuppression. Here, we report a new strategy for reconstructing the damaged corneal surface in a goat model of total limbal stem cell deficiency (LSCD) by autologous transplantation of epidermal adult stem cells (EpiASC). METHODS: EpiASC derived from adult goat ear skin by explant culture were purified by selecting single cell-derived clones. These EpiASC were cultivated on denuded human amniotic membrane (HAM) and transplanted onto goat eyes with total LSCD. The characteristics of both EpiASC and reconstructed corneal epithelium were identified by histology and immunohistochemistry. The clinical characteristic of reconstructed corneal surface was observed by digital camera. RESULTS: Ten LSCD goats (10 eyes) were treated with EpiASC transplantation, leading to the restoration of corneal transparency and improvement of postoperative visual acuity to varying degrees in 80.00% (8/10) of the experimental eyes. The corneal epithelium of control groups either with HAM transplantation only or without any transplantation showed irregular surfaces, diffuse vascularization, and pannus on the entire cornea. The reconstructed corneal epithelium (RCE) expressed CK3, CK12, and PAX-6 and had the function of secreting glycocalyx-like material (AB-PAS positive). During the follow-up period, all corneal surfaces remained transparent and there were no serious complications. We also observed that the REC expressed CK1/10 weakly at six months after operation but not at 12 months after operation, suggesting that the REC was derived from grafted EpiASC. CONCLUSIONS: Our results showed that EpiASC repaired the damaged cornea of goats with total LSCD and demonstrated that EpiASC can be induced to differentiate into corneal epithelial cell types in vivo, which at least in part correlated with down-regulation of CK1/10 and upregulation of PAX-6.


Assuntos
Córnea/patologia , Células Epidérmicas , Procedimentos de Cirurgia Plástica , Transplante de Células-Tronco , Âmnio/citologia , Animais , Substância Própria/patologia , Proteínas do Olho/metabolismo , Cabras , Humanos , Células-Tronco/citologia , Transplante Autólogo
11.
Tissue Eng ; 13(8): 2053-61, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17518714

RESUMO

The poorly vascularized fibrous capsule that develops around implantable biomedical devices (for drug delivery, biosensors, etc.) severely limits their applications. We tested the hypotheses that co-implantation of bone marrow-derived progenitor cells could stimulate the vascularization of implants. To assess the presence of functional peri-implant microvasculature, we developed a novel model of implanted device containing an oxygen (O(2))-sensing spin probe (detectable using electron paramagnetic resonance) placed inside a nanoporous filter-limited capsule. These devices were implanted subcutaneously in C57/Bl6 mice alone, with the addition of a Matrigel plug in front of the filter, or with the addition of Matrigel containing equal proportions of c-kit(+) and stem cell antigen-1(+) bone marrow-derived cells. Implants partial pressure of O(2) (pO(2)) were recorded non-invasively and periodically for up to 10 weeks. Tissue surrounding the implants was collected for immunohistochemistry. Initially, there were no differences in pO(2) between the experimental groups. After 3 weeks, the devices supplied with progenitor cells showed more than twice the O(2) concentrations as controls. This difference remained significant for 4 more weeks and then started to decrease slightly, still being 6 mmHg higher than in the controls at 10 weeks post-implantation. Collagen deposition was detected around the control implants, along with F4/80-positive macrophages and giant cells. In the plugs collected from the cell treatment group, we found an active process of adipogenesis, accompanied by neovascularization, and a highly vascularized adipose layer surrounding the implants. In conclusion, we successfully developed a cell therapy-type strategy to maintain vascularization around implanted devices using co-administration of bone marrow-derived progenitor cells, and we demonstrated a novel O(2)-sensing method to functionally monitor neovascularization in vivo.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Células-Tronco Hematopoéticas , Neovascularização Fisiológica , Oximetria , Animais , Antígenos Ly/biossíntese , Células da Medula Óssea/citologia , Células Cultivadas , Espectroscopia de Ressonância de Spin Eletrônica , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Pressão Parcial , Proteínas Proto-Oncogênicas c-kit/biossíntese
12.
Tissue Eng Part B Rev ; 23(3): 237-244, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27917703

RESUMO

Bioprinting is a technology with the prospect to change the way many diseases are treated, by replacing the damaged tissues with live de novo created biosimilar constructs. However, after more than a decade of incubation and many proofs of concept, the field is still in its infancy. The current stagnation is the consequence of its early success: the first bioprinters, and most of those that followed, were modified versions of the three-dimensional printers used in additive manufacturing, redesigned for layer-by-layer dispersion of biomaterials. In all variants (inkjet, microextrusion, or laser assisted), this approach is material ("scaffold") dependent and energy intensive, making it hardly compatible with some of the intended biological applications. Instead, the future of bioprinting may benefit from the use of gentler scaffold-free bioassembling methods. A substantial body of evidence has accumulated, indicating this is possible by use of preformed cell spheroids, which have been assembled in cartilage, bone, and cardiac muscle-like constructs. However, a commercial instrument capable to directly and precisely "print" spheroids has not been available until the invention of the microneedles-based ("Kenzan") spheroid assembling and the launching in Japan of a bioprinter based on this method. This robotic platform laces spheroids into predesigned contiguous structures with micron-level precision, using stainless steel microneedles ("kenzans") as temporary support. These constructs are further cultivated until the spheroids fuse into cellular aggregates and synthesize their own extracellular matrix, thus attaining the needed structural organization and robustness. This novel technology opens wide opportunities for bioengineering of tissues and organs.


Assuntos
Bioimpressão , Materiais Biocompatíveis , Procedimentos Cirúrgicos Robóticos , Esferoides Celulares , Engenharia Tecidual
13.
Biotechnol J ; 12(9)2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28731527

RESUMO

Immunomagnetic separation is used to isolate circulating endothelial cells (ECs) and endothelial progenitor cells (EPCs) for diagnostics and tissue engineering. However, potentially detrimental changes in cell properties have been observed post-separation. Here, the effect of mechanical force, which is naturally applied during immunomagnetic separation, on proliferation of human umbilical vein endothelial cells (HUVEC), kinase insert domain-positive receptor (KDR) cells, and peripheral blood mononuclear cells (PBMCs). Cells are exposed to CD31 or Vascular Endothelial Growth Factor Receptor-2 (VEGFR2) targeted MACSi beads at varying bead to cell ratios and compared to free antibody and unconjugated beads. A vertical magnetic gradient is applied to static 2D cultures, and a magnetic cell sorter is used to analyze cells in dynamic flow. No significant difference in EC proliferation is observed for controls or VEGFR2-targeting beads, whereas CD31-conjugated beads increase proliferation in a dose dependent manner in static 2-D cultures. This effect occurs in the absence of magnetic field, but is more pronounced with magnetic force. After flow sorting, similar increases in proliferation are seen for CD31 targeting beads. Thus, the effects of targeting antibody and magnetic force applied should be considered when designing immunomagnetic separation protocols for ECs.


Assuntos
Proliferação de Células/fisiologia , Células Endoteliais/fisiologia , Mecanotransdução Celular/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana , Humanos
14.
Biotechnol J ; 12(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29030959

RESUMO

Recently a protocol is established to obtain large quantities of human induced pluripotent stem cells (iPSC)-derived endothelial progenitors, called endothelial colony forming cells (ECFC), and of candidate smooth-muscle forming cells (SMFC). Here, the suitability for assembling in spheroids, and in larger 3D cell constructs is tested. iPSC-derived ECFC and SMFC are labeled with tdTomato and eGFP, respectively. Spheroids are formed in ultra-low adhesive wells, and their dynamic proprieties are studied by time-lapse microscopy, or by confocal microscopy. Spheroids are also tested for fusion ability either in the wells, or assembled on the Regenova 3D bioprinter which laces them in stainless steel micro-needles (the "Kenzan" method). It is found that both ECFC and SMFC formed spheroids in about 24 h. Fluorescence monitoring indicated a continuous compaction of ECFC spheroids, but stabilization in those prepared from SMFC. In mixed spheroids, the cell distribution changed continuously, with ECFC relocating to the core, and showing pre-vascular organization. All spheroids have the ability of in-well fusion, but only those containing SMFC are robust enough to sustain assembling in tubular structures. In these constructs a layered distribution of alpha smooth muscle actin-positive cells and extracellular matrix deposition is found. In conclusion, iPSC-derived vascular cell spheroids represent a promising new cellular material for scaffold-free biofabrication.


Assuntos
Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Esferoides Celulares/citologia , Engenharia Tecidual/métodos , Células Endoteliais da Veia Umbilical Humana , Humanos
16.
Circulation ; 106(9): 1154-8, 2002 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-12196344

RESUMO

BACKGROUND: Oxidant stress has been implicated in the pathogenesis of atherothrombosis and other vascular disorders accompanied by endothelial dysfunction. Glutathione peroxidases (GPx) play an important role in the cellular defense against oxidant stress by utilizing glutathione (GSH) to reduce lipid hydroperoxides and hydrogen peroxide to their corresponding alcohols. Cellular GPx (GPx-1) is the principal intracellular isoform of GPx. We hypothesized that GPx-1 deficiency per se induces endothelial dysfunction and structural vascular abnormalities through increased oxidant stress. METHODS AND RESULTS: A murine model of heterozygous deficiency of GPx-1 (GPx(+/-)) was investigated to examine this hypothesis. Mesenteric arterioles in GPx-1(+/-) mice demonstrated vasoconstriction to acetylcholine compared with vasodilation in wild-type mice (maximal change in vessel diameter, -13.0+/-2.8% versus 13.2+/-2.8%, P<0.0001). We also noted an increase in the plasma and aortic levels of the isoprostane iPF(2alpha)-III, a marker of oxidant stress, in GPx-1(+/-) mice compared with wild-type mice (170.4+/-23 pg/mL plasma versus 98.7+/-7.1 pg/mL plasma, P<0.03; 11.7+/-0.87 pg/mg aortic tissue versus 8.2+/-0.55 pg/mg aortic tissue, P<0.01). Histological sections from the coronary vasculature of GPx-1(+/-) mice show increased perivascular matrix deposition, an increase in the number of adventitial fibroblasts, and intimal thickening. These structural abnormalities in the myocardial vasculature were accompanied by diastolic dysfunction after ischemia-reperfusion. CONCLUSIONS: These findings demonstrate that heterozygous deficiency of GPx-1 leads to endothelial dysfunction, possibly associated with increased oxidant stress, and to significant structural vascular and cardiac abnormalities. These data illustrate the importance of this key antioxidant enzyme in functional and structural responses of the mammalian cardiovascular system.


Assuntos
Glutationa Peroxidase/deficiência , Mesentério/fisiopatologia , Erros Inatos do Metabolismo/fisiopatologia , Contração Miocárdica , Sistema Vasomotor/fisiopatologia , Animais , Antioxidantes/farmacologia , Aorta Torácica/química , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Bradicinina/farmacologia , Vasos Coronários/patologia , GMP Cíclico/análise , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Marcação de Genes , Glutationa Peroxidase/genética , Heterozigoto , Técnicas In Vitro , Masculino , Mesentério/irrigação sanguínea , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/patologia , Camundongos , Microcirculação/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/genética , Miocárdio/metabolismo , Miocárdio/patologia , Doadores de Óxido Nítrico/farmacologia , Estresse Oxidativo , Vasodilatadores/farmacologia , Sistema Vasomotor/efeitos dos fármacos , Glutationa Peroxidase GPX1
17.
Circulation ; 106(12): 1536-42, 2002 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-12234961

RESUMO

BACKGROUND: The killing of vascular cells by activated macrophages is an important step in the process of destabilization of the arterial wall. The death receptor Fas is implicated in vascular cell death. Hence, we extended our studies in a rat aortic allograft model, using adenovirus-mediated overexpression of soluble Fas (sFas) to block Fas binding to Fas ligand (Fas-L). The contribution of Fas to vascular cell injury and consequent transplant arteriosclerosis was investigated. METHODS AND RESULTS: Activated monocytes in the presence of macrophage colony-stimulating factor induce endothelial cell apoptosis in vitro, which was significantly inhibited by adenovirus-mediated sFas overexpression. Next, donor rat abdominal aortas were either untreated or transduced with adenoviruses encoding (1) rat soluble Fas (Ad3rsFas), (2) no insert (Ad3Null), and (3) beta-galactosidase (Ad3nBg). A total of 175 aortic grafts were harvested 2 to 90 days after transplantation. Vascular cell apoptosis and CD45+ cell infiltration were significantly reduced in Ad3rsFas-transduced aortas, as compared with control allografts. Moreover, the control allografts developed marked intimal thickening, whereas Ad3rsFas-transduced allografts had significantly less neointima until the 90-day time point. CONCLUSIONS: sFas overexpression protects the integrity of the vessel wall from immune injury and attenuates transplant arteriosclerosis.


Assuntos
Aorta/transplante , Arteriosclerose/prevenção & controle , Receptor fas/genética , Adenoviridae/genética , Animais , Aorta/citologia , Aorta/metabolismo , Apoptose , Arteriosclerose/etiologia , Arteriosclerose/patologia , Movimento Celular , Células Cultivadas , Endotélio Vascular/citologia , Vetores Genéticos , Humanos , Leucócitos/fisiologia , Masculino , Monócitos/fisiologia , RNA Mensageiro/biossíntese , Ratos , Transdução Genética , Receptor fas/metabolismo , Receptor fas/fisiologia
18.
Stem Cells Dev ; 14(2): 111-21, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15910238

RESUMO

There is increasing evidence that cells of disparate phenotypes displaying various degrees of proliferative capacity engraft and function heterotopically in adult organisms. Efforts were made to reconcile these findings with the embryologic notions of pluripotent stem or progenitor cell, although the nature of the 'stemness' remained elusive. This topic is particularly important for the cardiovascular system, in which cytotrophoblasts, certain tumor cells, monocytes/macrophages, peritoneal mesothelial cells, and others acquire endothelial properties and/or perform endothelial functions. Here we suggest that this pluripotency reflects a fundamental characteristic of cellular diversity, which is manifested as the adaptive response to a functional pressure exerted by the cell's biochemical and biophysical microenvironments that would drive their differentiation. In this model, differentiation is a dynamic, reversible, and open-ended process where the cells would maintain the flexibility to respond to changing environmental clues with a fine tuning of their structure, a property that was previously called cellular plasticity. Pluripotent adult stem cells that display this property in culture, and, perhaps upon in vivo administration, were described. Therefore, we also suggest that differentiation of stem cells is a form of cellular plasticity within the larger context of functional adaptation, whereas their stemness remains associated with self-renewal.


Assuntos
Sistema Cardiovascular/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco/citologia , Animais , Substitutos Sanguíneos , Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula , Endotélio Vascular/citologia , Humanos , Macrófagos/citologia , Monócitos/citologia , Neoplasias/patologia , Neovascularização Patológica , Peritônio/patologia , Células-Tronco Pluripotentes/citologia , Trofoblastos/citologia
19.
Thromb Haemost ; 93(2): 242-56, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15711739

RESUMO

For the past two decades much research on selective photothermolysis of port wine stain vasculature has been devoted to optimizing laser parameters. Unfortunately, 60% of patients still respond suboptimally to laser therapy, despite significant innovations in treatment strategies and laser technology. Here we present a novel treatment approach based on combining selective photothermolysis with the administration of prothrombotic and/or anti-fibrinolytic pharmaceutical agents, with the aim of enhancing vaso-occlusion and post-treatment remodelling in difficult-to-target vessels. A hypercoagulable state of blood will instill laser-induced occlusive thrombosis in a wider array of vessel diameters at greater dermal depths, whereby larger vascular segments will ultimately undergo the chronic inflammatory processes that result in blood volume reduction, and thus lesional blanching. With thrombosis as a primary trigger for these inflammatory processes, we have extrapolated the threshold damage profile that is required for clinically relevant thrombus formation. Consequently, a recently proposed model of thrombus organization, in which recanalization is associated with endothelial progenitor cell-mediated neovasculogenesis, is elaborated in the framework of lesional blanching and juxtaposed to angiogenic reconstruction of affected dermal vasculature. Since neovasculogenesis and angiogenesis are regulated by the degree of vaso-occlusion and corollary drop in local oxygen tension, both can be manipulated by the administration of procoagulant pharmaceuticals. Lastly, in an effort to optimally balance selective photothermolysis with pharmacokinetics and clinical safety, the use of a gold nanoshell drug delivery system, in which the procoagulant drugs are encapsulated by a wavelength-modulated, gold-coated polymer matrix, is proposed. We have termed this modality site-specific pharmaco-laser therapy.


Assuntos
Coagulantes/uso terapêutico , Terapia a Laser , Fotólise , Mancha Vinho do Porto/terapia , Adjuvantes Farmacêuticos , Coagulantes/administração & dosagem , Humanos , Neovascularização Fisiológica , Trombose/etiologia
20.
Trends Cardiovasc Med ; 13(7): 265-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14522465

RESUMO

Recent experimental data suggest a role for blood-borne mononuclear cells, including bone marrow-derived endothelial progenitor cells (EPCs) in organization, vascularization, and recanalization of thrombi. Older studies have described in detail the in situ structural modifications accompanying these processes. The common themes are (a) involvement of pluripotent mononuclear cells, and (b) proteolytic and/or phagocytic activity of monocytes/macrophages (MCs/Mphs) in modifying the thrombus to a proangiogenic state. In addition to a nurturing function, MCs/Mphs may assist the engraftment of thrombus-trapped or incoming EPCs. The differences between the recanalization potency of venous and arterial thrombi suggest that in addition to the organization of the fibrin meshwork, the cellular composition of the venous thrombi may be responsible for the better recanalization seen in venous clots. These observations set the stage for therapeutic manipulation of organization and recanalization of thrombi, by increasing the MC/Mph and/or EPC content.


Assuntos
Monócitos/fisiologia , Neovascularização Patológica/fisiopatologia , Trombose/fisiopatologia , Animais , Quimiocina CCL2/fisiologia , Células Endoteliais/fisiologia , Humanos , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA