Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bio Protoc ; 14(2): e4919, 2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-38268973

RESUMO

Human skin reconstruction on immune-deficient mice has become indispensable for in vivo studies performed in basic research and translational laboratories. Further advancements in making sustainable, prolonged skin equivalents to study new therapeutic interventions rely on reproducible models utilizing patient-derived cells and natural three-dimensional culture conditions mimicking the structure of living skin. Here, we present a novel step-by-step protocol for grafting human skin cells onto immunocompromised mice that requires low starting cell numbers, which is essential when primary patient cells are limited for modeling skin conditions. The core elements of our method are the sequential transplantation of fibroblasts followed by keratinocytes seeded into a fibrin-based hydrogel in a silicone chamber. We optimized the fibrin gel formulation, timing for gel polymerization in vivo, cell culture conditions, and seeding density to make a robust and efficient grafting protocol. Using this approach, we can successfully engraft as few as 1.0 × 106 fresh and 2.0 × 106 frozen-then-thawed keratinocytes per 1.4 cm2 of the wound area. Additionally, it was concluded that a successful layer-by-layer engraftment of skin cells in vivo could be obtained without labor-intensive and costly methodologies such as bioprinting or engineering complex skin equivalents. Key features • Expands upon the conventional skin chamber assay method (Wang et al., 2000) to generate high-quality skin grafts using a minimal number of cultured skin cells. • The proposed approach allows the use of frozen-then-thawed keratinocytes and fibroblasts in surgical procedures. • This system holds promise for evaluating the functionality of skin cells derived from induced pluripotent stem cells and replicating various skin phenotypes. • The entire process, from thawing skin cells to establishing the graft, requires 54 days. Graphical overview.

2.
Methods Mol Biol ; 2549: 153-167, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33772462

RESUMO

Differentiating human induced pluripotent stem cells (iPSCs) into multipotent mesenchymal stem/stromal cells (MSCs) offers a renewable source of therapeutically invaluable cells. However, the process of MSC derivation from iPSCs suffers from an undesirably low efficiency. In this chapter, we present an optimized procedure to produce MSCs from human iPSCs with a high efficiency. The protocol depends on the generation of embryoid bodies (EBs) and requires the treatment of EBs with transforming growth factor beta 1 (TGF-ß1). The resulting MSCs can be purified based on the expression of CD73, CD105, and CD90 markers and expanded for multiple passages without losing their characteristics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Diferenciação Celular , Corpos Embrioides/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Antígenos Thy-1/metabolismo
3.
Methods Mol Biol ; 2549: 169-186, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33755906

RESUMO

Endothelial cells (ECs) are important components of the circulatory system. These cells can be used for in vitro modeling of cardiovascular diseases and in regenerative medicine to promote vascularization of engineered tissue constructs. However, low proliferative capacity and patient-to-patient variability limit the use of primary ECs in the clinic and disease modeling. ECs differentiated from human induced pluripotent stem cells (iPSCs) can serve as a viable alternative to primary ECs for these applications. This is because human iPSCs can proliferate indefinitely and have the potential to differentiate into a variety of somatic cell lines, providing a renewable source of patient-specific cells. Here, we present an optimized, highly reproducible method for the differentiation of human iPSCs toward vascular ECs. The protocol relies on the activation of the WNT signaling pathway and the use of growth factors and small molecules. The resulting iPSC-derived ECs can be cultured for multiple passages without losing their functionality and are suitable for both in vitro and in vivo studies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular/fisiologia , Linhagem Celular , Células Endoteliais/metabolismo , Humanos , Medicina Regenerativa
4.
Am J Med Genet A ; 185(11): 3390-3400, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34435747

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare genodermatosis caused by mutations in the gene coding for type VII collagen (COL7A1). More than 800 different pathogenic mutations in COL7A1 have been described to date; however, the ancestral origins of many of these mutations have not been precisely identified. In this study, 32 RDEB patient samples from the Southwestern United States, Mexico, Chile, and Colombia carrying common mutations in the COL7A1 gene were investigated to determine the origins of these mutations and the extent to which shared ancestry contributes to disease prevalence. The results demonstrate both shared European and American origins of RDEB mutations in distinct populations in the Americas and suggest the influence of Sephardic ancestry in at least some RDEB mutations of European origins. Knowledge of ancestry and relatedness among RDEB patient populations will be crucial for the development of future clinical trials and the advancement of novel therapeutics.


Assuntos
Colágeno Tipo VII/genética , Epidermólise Bolhosa Distrófica/genética , Hispânico ou Latino/genética , Judeus/genética , Chile/epidemiologia , Colômbia/epidemiologia , Epidermólise Bolhosa Distrófica/epidemiologia , Feminino , Genes Recessivos/genética , Humanos , Masculino , México/epidemiologia , Fenótipo , Estados Unidos/epidemiologia
5.
Membranes (Basel) ; 11(7)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203550

RESUMO

The aim of this work is to develop a new type of carbon-ceramic membranes for the removal of pharmaceutical substances from water. The membranes were prepared by the chemical modification method using an organosilicon precursor-octadecyltrichlorosilane (ODTS). Graphene oxide, multi-walled carbon nanotubes with carboxylic groups, and single-walled carbon nanotubes were used in the modification process. The filtration properties and adsorption properties of the developed membranes were tested. In order to characterize the membrane, the water permeability, the change of the permeate flux in time, and the adsorbed mass of the substance were determined. Additionally, the surface properties of the membranes were characterized by contact angle measurements and porosimetry. The antibiotic tetracycline was used in the adsorption tests. Based on the results, the improved adsorption properties of the modified membrane in relation to the unmodified membrane were noticed. Novel ceramic membranes modified with MWCNT are characterized by 45.4% removal of tetracycline and permeate flux of 520 L·h·m-2·bar-1. We demonstrated the ability of modified membranes to adsorb pharmaceuticals from water streams that are in contact with the membrane. Novel membranes retain their filtration properties. Therefore, such membranes can be used in an integrated filtration-adsorption process.

6.
Methods Mol Biol ; 2155: 11-21, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32474864

RESUMO

The discovery of induced pluripotent stem cell (iPSC) technology has provided a versatile platform for basic science research and regenerative medicine. With the rise of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) systems and the ease at which they can be utilized for gene editing, creating genetically modified iPSCs has never been more advantageous for studying both organism development and potential clinical applications. However, to better understand the behavior and true therapeutic potential of iPSCs and iPSC-derived cells, a tool for labeling and monitoring these cells in vitro and in vivo is needed. Here, we describe a protocol that provides a straightforward method for introducing a stable, highly expressed fluorescent protein into iPSCs using the CRISPR/Cas9 system and a standardized donor vector. The approach involves the integration of the EGFP transgene into the transcriptionally active adeno-associated virus integration site 1 (AAVS1) locus through homology directed repair. The knockin of this transgene results in the generation of iPSC lines with constitutive expression of the EGFP protein that also persists in differentiated iPSCs. These EGFP-labeled iPSC lines are ideal for assessing iPSC differentiation in vitro and evaluating the distribution of iPSC-derived cells in vivo after transplantation into model animals.


Assuntos
Expressão Gênica , Genes Reporter , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Sistemas CRISPR-Cas , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Edição de Genes , Técnicas de Introdução de Genes , Marcação de Genes , Engenharia Genética/métodos , Vetores Genéticos/genética , Humanos
7.
Methods Mol Biol ; 2117: 271-284, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31960386

RESUMO

Reprogramming a patient's somatic cells into induced pluripotent stem cells (iPSCs) holds great promise for disease modeling and the development of autologous cellular therapeutics. However, it remains challenging to consistently reprogram primary human cells, as they are frequently aged, diseased, or in low abundance. Here we present a modified highly efficient and clinically relevant RNA-based method for reprogramming disease-associated and other difficult-to-reprogram human primary fibroblast lines into iPSCs. We also describe optimizations that can be employed for consistent reprogramming of these difficult-to-reprogram cells. With the provided protocol, integration-free iPSC lines can be successfully generated from a small number of primary human fibroblasts in approximately 5-7 weeks.


Assuntos
Técnicas de Reprogramação Celular/métodos , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Cultura Primária de Células/métodos , RNA/genética , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Humanos
8.
Methods Mol Biol ; 2109: 169-183, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31119714

RESUMO

Human skin equivalents composed of epidermal cells and fibroblasts are important for modeling human epidermal development, testing new therapeutics, and designing novel treatment strategies for human skin diseases. Here, we describe a procedure for the generation of an in vivo full-thickness human skin equivalent on an immunodeficient mouse using a grafting chamber system. The protocol involves mixing human epidermal cells and fibroblasts in a silicone grafting chamber that is surgically inserted onto the muscle fascia of a recipient immunodeficient mouse. Following the removal of the silicone chamber, the graft area is exposed to air to induce stratification of developing epidermis, resulting in the reconstitution of full-thickness human skin tissue on a live mouse. This grafting system provides a straightforward approach to study human skin diseases in an animal model and has been previously used to determine the ability of both mouse and human primary epidermal cells and cells derived from pluripotent stem cells to regenerate functional skin in vivo.


Assuntos
Fibroblastos/citologia , Queratinócitos/citologia , Transplante de Pele/métodos , Animais , Células Cultivadas , Humanos , Hospedeiro Imunocomprometido , Fígado/cirurgia , Camundongos , Camundongos Endogâmicos NOD , Modelos Animais , Cultura Primária de Células , Técnicas de Cultura de Tecidos
9.
J Vis Exp ; (141)2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30531717

RESUMO

Induced pluripotent stem cells (iPSCs) have proven to be a valuable tool to study human development and disease. Further advancing iPSCs as a regenerative therapeutic requires a safe, robust, and expedient reprogramming protocol. Here, we present a clinically relevant, step-by-step protocol for the extremely high-efficiency reprogramming of human dermal fibroblasts into iPSCs using a non-integrating approach. The core of the protocol consists of expressing pluripotency factors (SOX2, KLF4, cMYC, LIN28A, NANOG, OCT4-MyoD fusion) from in vitro transcribed messenger RNAs synthesized with modified nucleotides (modified mRNAs). The reprogramming modified mRNAs are transfected into primary fibroblasts every 48 h together with mature embryonic stem cell-specific microRNA-367/302 mimics for two weeks. The resulting iPSC colonies can then be isolated and directly expanded in feeder-free conditions. To maximize efficiency and consistency of our reprogramming protocol across fibroblast samples, we have optimized various parameters including the RNA transfection regimen, timing of transfections, culture conditions, and seeding densities. Importantly, our method generates high-quality iPSCs from most fibroblast sources, including difficult-to-reprogram diseased, aged, and/or senescent samples.


Assuntos
Reprogramação Celular/fisiologia , Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , MicroRNAs/fisiologia , RNA Mensageiro/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Engenharia Genética/métodos , Humanos , Fator 4 Semelhante a Kruppel , Transfecção/métodos
10.
J Biol Chem ; 293(47): 18309-18317, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30275014

RESUMO

Sec1/Munc18 (SM) proteins promote intracellular vesicle fusion by binding to N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). A key SNARE-binding mode of SM proteins involves the N-terminal peptide (N-peptide) motif of syntaxin, a SNARE subunit localized to the target membrane. In in vitro membrane fusion assays, inhibition of N-peptide motif binding previously has been shown to abrogate the stimulatory function of Munc18-1, a SM protein involved in synaptic exocytosis in neurons. The physiological role of the N-peptide-binding mode, however, remains unclear. In this work, we addressed this key question using a "clogged" Munc18-1 protein, in which an ectopic copy of the syntaxin N-peptide motif was directly fused to Munc18-1. We found that the ectopic N-peptide motif blocks the N-peptide-binding pocket of Munc18-1, preventing the latter from binding to the native N-peptide motif on syntaxin-1. In a reconstituted system, we observed that clogged Munc18-1 is defective in promoting SNARE zippering. When introduced into induced neuronal cells (iN cells) derived from human pluripotent stem cells, clogged Munc18-1 failed to mediate synaptic exocytosis. As a result, both spontaneous and evoked synaptic transmission was abolished. These genetic findings provide direct evidence for the crucial role of the N-peptide-binding mode of Munc18-1 in synaptic exocytosis. We suggest that clogged SM proteins will also be instrumental in defining the physiological roles of the N-peptide-binding mode in other vesicle-fusion pathways.


Assuntos
Exocitose , Proteínas Munc18/química , Proteínas Munc18/metabolismo , Peptídeos/metabolismo , Sinapses/metabolismo , Motivos de Aminoácidos , Humanos , Proteínas Munc18/genética , Neurônios/química , Neurônios/metabolismo , Peptídeos/química , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Sinapses/química , Sinapses/genética , Transmissão Sináptica , Sintaxina 1/química , Sintaxina 1/genética , Sintaxina 1/metabolismo
11.
Nat Commun ; 9(1): 745, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467427

RESUMO

Induced pluripotent stem cells (iPSCs) hold great promise for regenerative medicine; however, their potential clinical application is hampered by the low efficiency of somatic cell reprogramming. Here, we show that the synergistic activity of synthetic modified mRNAs encoding reprogramming factors and miRNA-367/302s delivered as mature miRNA mimics greatly enhances the reprogramming of human primary fibroblasts into iPSCs. This synergistic activity is dependent upon an optimal RNA transfection regimen and culturing conditions tailored specifically to human primary fibroblasts. As a result, we can now generate up to 4,019 iPSC colonies from only 500 starting human primary neonatal fibroblasts and reprogram up to 90.7% of individually plated cells, producing multiple sister colonies. This methodology consistently generates clinically relevant, integration-free iPSCs from a variety of human patient's fibroblasts under feeder-free conditions and can be applicable for the clinical translation of iPSCs and studying the biology of reprogramming.


Assuntos
Técnicas de Reprogramação Celular , Linhagem Celular , Fibroblastos , Humanos , Células-Tronco Pluripotentes Induzidas , RNA
12.
Methods Mol Biol ; 1195: 1-12, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24510784

RESUMO

Direct reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) provides an opportunity to develop novel personalized treatment options for numerous diseases and to advance current approaches for cell-based drug discoveries and disease modeling. The ability to differentiate iPSCs into relevant cell types is an important prerequisite for the successful development of iPSC-based treatment and modeling strategies. Here, we describe a protocol for the efficient differentiation of human iPSCs into functional keratinocytes. The protocol employs treating iPSCs with retinoic acid and bone-morphogenetic protein-4 to induce differentiation toward a keratinocyte lineage, which is then followed by the growth of differentiated iPSCs on collagen type I- and collagen type IV-coated dishes to enrich for iPSC-derived keratinocytes.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Queratinócitos/citologia , Proteína Morfogenética Óssea 4/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Colágeno Tipo I/farmacologia , Colágeno Tipo IV/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Tretinoína/farmacologia
13.
J Immunol ; 189(6): 2965-74, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22904300

RESUMO

The majority of the human population becomes infected early in life by the gammaherpesvirus EBV. Some findings suggest that there is an association between EBV infection and the appearance of pathogenic Abs found in lupus. Gammaherpesvirus 68 infection of adult mice (an EBV model) was shown to induce polyclonal B cell activation and hypergammaglobulinemia, as well as increased production of autoantibodies. In this study, we explored the possibility that this breach of tolerance reflects loss of B cell anergy. Our findings show that, although anergic B cells transiently acquire an activated phenotype early during infection, they do not become responsive to autoantigen, as measured by the ability to mobilize Ca2+ following AgR cross-linking or mount Ab responses following immunization. Indeed, naive B cells also acquire an activated phenotype during acute infection but are unable to mount Ab responses to either T cell-dependent or T cell-independent Ags. In acutely infected animals, Ag stimulation leads to upregulation of costimulatory molecules and relocalization of Ag-specific B cells to the B-T cell border; however, these cells do not proliferate or differentiate into Ab-secreting cells. Adoptive-transfer experiments show that the suppressed state is reversible and is dictated by the environment in the infected host. Finally, B cells in infected mice deficient of CD4+ T cells are not suppressed, suggesting a role for CD4+ T cells in enforcing unresponsiveness. Thus, rather than promoting loss of tolerance, gammaherpesvirus 68 infection induces an immunosuppressed state, reminiscent of compensatory anti-inflammatory response syndrome.


Assuntos
Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Subpopulações de Linfócitos B/imunologia , Anergia Clonal/imunologia , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Tolerância Imunológica , Doença Aguda , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Subpopulações de Linfócitos B/patologia , Anergia Clonal/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Tolerância Imunológica/genética , Imunofenotipagem , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
14.
Semin Immunol ; 24(5): 342-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22560930

RESUMO

In this review we discuss the changes that occur in the B lymphocyte compartment of mice and humans as they progress to old age, focusing on recent advances in this important area of research. Primary areas considered include increased morbidity and mortality in the elderly following infection, and decreased responsiveness to vaccines that evoke primary humoral immune responses, as well as those that evoke responses by memory B cells generated following vaccination and natural infection earlier in life. We then consider some of the mechanisms that may underlie these observed declines in humoral immune function. This includes a discussion of alterations in B cell repertoire and subcompartment distribution, as well as defects in B lymphopoiesis, cell development and homeostasis that may contribute to these alterations, and ultimately to declining protective quality of antibodies produced in the elderly.


Assuntos
Envelhecimento , Linfócitos B/imunologia , Animais , Linfócitos B/citologia , Proliferação de Células , Homeostase , Humanos , Linfopoese , Vacinação
15.
Genes Dev ; 23(19): 2345-57, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19797771

RESUMO

Cohesins mediate sister chromatid cohesion and DNA repair and also function in gene regulation. Chromosomal cohesins are distributed nonrandomly, and their deposition requires the heterodimeric Scc2/Scc4 loader. Whether Scc2/Scc4 establishes nonrandom cohesin distributions on chromosomes is poorly characterized, however. To better understand the spatial regulation of cohesin association, we mapped budding yeast Scc2 and Scc4 chromosomal distributions. We find that Scc2/Scc4 resides at previously mapped cohesin-associated regions (CARs) in pericentromeric and arm regions, and that Scc2/Scc4-cohesin colocalization persists after the initial deposition of cohesins in G1/S phase. Pericentromeric Scc2/Scc4 enrichment is kinetochore-dependent, and both Scc2/Scc4 and cohesin associations are coordinately reduced in these regions following chromosome biorientation. Thus, these characteristics of Scc2/Scc4 binding closely recapitulate those of cohesin. Although present in G1, Scc2/Scc4 initially has a poor affinity for CARs, but its affinity increases as cells traverse the cell cycle. Scc2/Scc4 association with CARs is independent of cohesin, however. Taken together, these observations are inconsistent with a previous suggestion that cohesins are relocated by translocating RNA polymerases from separate loading sites to intergenic regions between convergently transcribed genes. Rather, our findings suggest that budding yeast cohesins are targeted to CARs largely by Scc2/Scc4 loader association at these locations.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos Fúngicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ciclo Celular/fisiologia , Cromatina/metabolismo , Cinetocoros/metabolismo , Coesinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...