Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1226615, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37842306

RESUMO

Background: Diabetes mellitus is characterized by chronic hyperglycemia with loss of ß-cell function and mass. An attractive therapeutic approach to treat patients with diabetes in a non-invasive way is to harness the innate regenerative potential of the pancreas. The Islet Neogenesis-Associated Protein pentadecapeptide (INGAP-PP) has been shown to induce ß-cell regeneration and improve their function in rodents. To investigate its possible mechanism of action, we report here the global transcriptional effects induced by the short-term INGAP-PP in vitro treatment of adult rat pancreatic islets. Methods and findings: Rat pancreatic islets were cultured in vitro in the presence of INGAP-PP for 4 days, and RNA-seq was generated from triplicate treated and control islet samples. We performed a de novo rat gene annotation based on the alignment of RNA-seq reads. The list of INGAP-PP-regulated genes was integrated with epigenomic data. Using the new gene annotation generated in this work, we quantified RNA-seq data profiled in INS-1 cells treated with IL1ß, IL1ß+Calcipotriol (a vitamin D agonist) or vehicle, and single-cell RNA-seq data profiled in rat pancreatic islets. We found 1,669 differentially expressed genes by INGAP-PP treatment, including dozens of previously unannotated rat transcripts. Genes differentially expressed by the INGAP-PP treatment included a subset of upregulated transcripts that are associated with vitamin D receptor activation. Supported by epigenomic and single-cell RNA-seq data, we identified 9 previously unannotated long noncoding RNAs (lncRNAs) upregulated by INGAP-PP, some of which are also differentially regulated by IL1ß and vitamin D in ß-cells. These include Ri-lnc1, which is enriched in mature ß-cells. Conclusions: Our results reveal the transcriptional program that could explain the enhancement of INGAP-PP-mediated physiological effects on ß-cell mass and function. We identified novel lncRNAs that are induced by INGAP-PP in rat islets, some of which are selectively expressed in pancreatic ß-cells and downregulated by IL1ß treatment of INS-1 cells. Our results suggest a relevant function for Ri-lnc1 in ß-cells. These findings are expected to provide the basis for a deeper understanding of islet translational results from rodents to humans, with the ultimate goal of designing new therapies for people with diabetes.


Assuntos
Diabetes Mellitus , Ilhotas Pancreáticas , RNA Longo não Codificante , Ratos , Humanos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Associadas a Pancreatite/genética , Proteínas Associadas a Pancreatite/metabolismo , Proteínas Associadas a Pancreatite/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Peptídeos/metabolismo , Diabetes Mellitus/metabolismo , Vitamina D/metabolismo
2.
BMC Genomics ; 22(1): 585, 2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34340653

RESUMO

BACKGROUND: Loss of pancreatic insulin-secreting ß-cells due to metabolic or autoimmune damage leads to the development of diabetes. The discovery that α-cells can be efficiently reprogrammed into insulin-secreting cells in mice and humans has opened promising avenues for innovative diabetes therapies. ß-cell loss triggers spontaneous reprogramming of only 1-2% of α-cells, limiting the extent of regeneration. Most α-cells are refractory to conversion and their global transcriptomic response to severe ß-cell loss as well as the mechanisms opposing their reprogramming into insulin producers are largely unknown. Here, we performed RNA-seq on FAC-sorted α-cells to characterize their global transcriptional responses at different time points after massive ß-cell ablation. RESULTS: Our results show that α-cells undergo stage-specific transcriptional changes 5- and 15-days post-diphtheria toxin (DT)-mediated ß-cell ablation. At 5 days, α-cells transiently upregulate various genes associated with interferon signaling and proliferation, including Interferon Induced Protein with Tetratricopeptide Repeats 3 (Ifit3). Subsequently, at 15 days post ß-cell ablation, α-cells undergo a transient downregulation of genes from several pathways including Insulin receptor, mTOR and MET signaling. CONCLUSIONS: The results presented here pinpoint novel markers discriminating α-cells at different stages after acute ß-cell loss, and highlight additional signaling pathways that are modulated in α-cells in this context.


Assuntos
Diabetes Mellitus , Células Secretoras de Glucagon , Células Secretoras de Insulina , Animais , Insulina , Camundongos , Transcriptoma
3.
Stem Cell Reports ; 16(9): 2289-2304, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34450036

RESUMO

Heterozygous mutations in HNF1B in humans result in a multisystem disorder, including pancreatic hypoplasia and diabetes mellitus. Here we used a well-controlled human induced pluripotent stem cell pancreatic differentiation model to elucidate the molecular mechanisms underlying HNF1B-associated diabetes. Our results show that lack of HNF1B blocks specification of pancreatic fate from the foregut progenitor (FP) stage, but HNF1B haploinsufficiency allows differentiation of multipotent pancreatic progenitor cells (MPCs) and insulin-secreting ß-like cells. We show that HNF1B haploinsufficiency impairs cell proliferation in FPs and MPCs. This could be attributed to impaired induction of key pancreatic developmental genes, including SOX11, ROBO2, and additional TEAD1 target genes whose function is associated with MPC self-renewal. In this work we uncover an exhaustive list of potential HNF1B gene targets during human pancreas organogenesis whose downregulation might underlie HNF1B-associated diabetes onset in humans, thus providing an important resource to understand the pathogenesis of this disease.


Assuntos
Diferenciação Celular/genética , Fator 1-beta Nuclear de Hepatócito/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Organogênese/genética , Pâncreas/embriologia , Pâncreas/metabolismo , Biomarcadores , Sistemas CRISPR-Cas , Linhagem da Célula/genética , Diabetes Mellitus/etiologia , Suscetibilidade a Doenças , Imunofluorescência , Edição de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Imunofenotipagem , Células Secretoras de Insulina/metabolismo , Transdução de Sinais
4.
Front Endocrinol (Lausanne) ; 12: 692596, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34295307

RESUMO

The occurrence of diabetes mellitus is characterized by pancreatic ß cell loss and chronic hyperglycemia. While Type 1 and Type 2 diabetes are the most common types, rarer forms involve mutations affecting a single gene. This characteristic has made monogenic diabetes an interesting disease group to model in vitro using human pluripotent stem cells (hPSCs). By altering the genotype of the original hPSCs or by deriving human induced pluripotent stem cells (hiPSCs) from patients with monogenic diabetes, changes in the outcome of the in vitro differentiation protocol can be analyzed in detail to infer the regulatory mechanisms affected by the disease-associated genes. This approach has been so far applied to a diversity of genes/diseases and uncovered new mechanisms. The focus of the present review is to discuss the latest findings obtained by modeling monogenic diabetes using hPSC-derived pancreatic cells generated in vitro. We will specifically focus on the interpretation of these studies, the advantages and limitations of the models used, and the future perspectives for improvement.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina/citologia , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Humanos , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento
5.
J Cell Biol ; 219(9)2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32673398

RESUMO

In mammals, argonaute (AGO) proteins have been characterized for their roles in small RNA-mediated posttranscriptional and also in transcriptional gene silencing. Here, we report a different role for AGO1 in estradiol-triggered transcriptional activation in human cells. We show that in MCF-7 mammary gland cells, AGO1 associates with transcriptional enhancers of estrogen receptor α (ERα) and that this association is up-regulated by treating the cells with estrogen (E2), displaying a positive correlation with the activation of these enhancers. Moreover, we show that AGO1 interacts with ERα and that this interaction is also increased by E2 treatment, but occurs in the absence of RNA. We show that AGO1 acts positively as a coactivator in estradiol-triggered transcription regulation by promoting ERα binding to its enhancers. Consistently, AGO1 depletion decreases long-range contacts between ERα enhancers and their target promoters. Our results point to a role of AGO1 in transcriptional regulation in human cells that is independent from small RNA binding.


Assuntos
Proteínas Argonautas/genética , Estrogênios/genética , Fatores de Iniciação em Eucariotos/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Linhagem Celular , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos/genética , Estradiol/genética , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Células MCF-7 , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética
6.
Nucleic Acids Res ; 47(20): 10645-10661, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31598691

RESUMO

The glucocorticoid and progesterone receptors (GR and PR) are closely related members of the steroid receptor family. Despite sharing similar structural and functional characteristics; the cognate hormones display very distinct physiological responses. In mammary epithelial cells, PR activation is associated with the incidence and progression of breast cancer, whereas the GR is related to growth suppression and differentiation. Despite their pharmacological relevance, only a few studies have compared GR and PR activities in the same system. Using a PR+/GR+ breast cancer cell line, here we report that either glucocorticoid-free or dexamethasone (DEX)-activated GR inhibits progestin-dependent gene expression associated to epithelial-mesenchymal-transition and cell proliferation. When both receptors are activated with their cognate hormones, PR and GR can form part of the same complex according to co-immunoprecipitation, quantitative microscopy and sequential ChIP experiments. Moreover, genome-wide studies in cells treated with either DEX or R5020, revealed the presence of several regions co-bound by both receptors. Surprisingly, GR also binds novel genomic sites in cells treated with R5020 alone. This progestin-induced GR binding was enriched in REL DNA motifs and located close to genes coding for chromatin remodelers. Understanding GR behavior in the context of progestin-dependent breast cancer could provide new targets for tumor therapy.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Sequência de Bases , Sítios de Ligação , Neoplasias da Mama/patologia , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cromatina/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Progestinas/farmacologia , Promegestona/farmacologia , Ligação Proteica/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
7.
Endocr Relat Cancer ; 26(1): 13-29, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30121620

RESUMO

Preclinical and clinical studies support that Notch signaling may play an important oncogenic role in cancer, but there is scarce information for pituitary tumors. We therefore undertook a functional study to evaluate Notch participation in pituitary adenoma growth. Tumors generated in Nude mice by subcutaneous GH3 somatolactotrope cell injection were treated in vivo with DAPT, a γ-secretase inhibitor, thus inactivating Notch signaling. This treatment led to pituitary tumor reduction, lower prolactin and GH tumor content and a decrease in angiogenesis. Furthermore, in silico transcriptomic and epigenomic analyses uncovered several tumor suppressor genes related to Notch signaling in pituitary tissue, namely Btg2, Nr4a1, Men1, Zfp36 and Cnot1. Gene evaluation suggested that Btg2, Nr4a1 and Cnot1 may be possible players in GH3 xenograft growth. Btg2 mRNA expression was lower in GH3 tumors compared to the parental line, and DAPT increased its expression levels in the tumor in parallel with the inhibition of its volume. Cnot1 mRNA levels were also increased in the pituitary xenografts by DAPT treatment. And the Nr4a1 gene was lower in tumors compared to the parental line, though not modified by DAPT. Finally, because DAPT in vivo may also be acting on tumor microenvironment, we determined the direct effect of DAPT on GH3 cells in vitro. We found that DAPT decreases the proliferative, secretory and migration potential of GH3 cells. These results position selective interruption of Notch signaling as a potential therapeutic tool in adjuvant treatments for aggressive or resistant pituitary tumors.


Assuntos
Adenoma/patologia , Neoplasias Hipofisárias/patologia , Receptores Notch/antagonistas & inibidores , Adenoma/metabolismo , Animais , Linhagem Celular Tumoral , Diaminas/farmacologia , Feminino , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Hipofisárias/metabolismo , Prolactina/metabolismo , Ratos , Receptores Notch/metabolismo , Transdução de Sinais , Tiazóis/farmacologia , Carga Tumoral
9.
Nat Cell Biol ; 17(5): 615-626, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25915126

RESUMO

The genomic regulatory programmes that underlie human organogenesis are poorly understood. Pancreas development, in particular, has pivotal implications for pancreatic regeneration, cancer and diabetes. We have now characterized the regulatory landscape of embryonic multipotent progenitor cells that give rise to all pancreatic epithelial lineages. Using human embryonic pancreas and embryonic-stem-cell-derived progenitors we identify stage-specific transcripts and associated enhancers, many of which are co-occupied by transcription factors that are essential for pancreas development. We further show that TEAD1, a Hippo signalling effector, is an integral component of the transcription factor combinatorial code of pancreatic progenitor enhancers. TEAD and its coactivator YAP activate key pancreatic signalling mediators and transcription factors, and regulate the expansion of pancreatic progenitors. This work therefore uncovers a central role for TEAD and YAP as signal-responsive regulators of multipotent pancreatic progenitors, and provides a resource for the study of embryonic development of the human pancreas.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Multipotentes/metabolismo , Proteínas Nucleares/metabolismo , Pâncreas/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Biologia Computacional , Proteínas de Ligação a DNA/genética , Bases de Dados Genéticas , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Organogênese , Pâncreas/embriologia , Fenótipo , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição de Domínio TEA , Fatores de Tempo , Fatores de Transcrição/genética , Proteínas de Sinalização YAP , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
10.
Clin Cancer Res ; 20(15): 4086-95, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24842953

RESUMO

PURPOSE: Clinical and epidemiologic data suggest that obesity is associated with more aggressive forms of prostate cancer, poor prognosis, and increased mortality. C-terminal-binding protein 1 (CtBP1) is a transcription repressor of tumor suppressor genes and is activated by NADH binding. High calorie intake decreases intracellular NAD(+)/NADH ratio. The aim of this work was to assess the effect of high-fat diet (HFD) and CtBP1 expression modulation over prostate xenograft growth. EXPERIMENTAL DESIGN: We developed a metabolic syndrome-like disease in vivo model by feeding male nude mice with HFD during 16 weeks. Control diet (CD)-fed animals were maintained at the same conditions. Mice were inoculated with PC3 cells stable transfected with shCtBP1 or control plasmids. Genome-wide expression profiles and Gene Set Enrichment Analysis (GSEA) were performed from PC3.shCtBP1 versus PC3.pGIPZ HFD-fed mice tumors. RESULTS: No significant differences were observed in tumor growth on CD-fed mice; however, we found that only 60% of HFD-fed mice inoculated with CtBP1-depleted cells developed a tumor. Moreover these tumors were significantly smaller than those generated by PC3.pGIPZ control xenografts. We found 823 genes differentially expressed in shCtBP1 tumors from HFD-fed mice. GSEA from expression dataset showed that most of these genes correspond to cell adhesion, metabolic process, and cell cycle. CONCLUSIONS: Metabolic syndrome-like diseases and CtBP1 expression cooperate to induce prostate tumor growth. Hence, targeting of CtBP1 expression might be considered for prostate cancer management and therapy in the subset of patients with metabolic syndromes.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Dieta Hiperlipídica/efeitos adversos , Síndrome Metabólica/prevenção & controle , Obesidade/prevenção & controle , Neoplasias da Próstata/prevenção & controle , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Adesão Celular , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Hormônios Esteroides Gonadais/farmacologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/patologia , Camundongos , Camundongos Nus , Obesidade/etiologia , Obesidade/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Genet ; 46(2): 136-143, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24413736

RESUMO

Type 2 diabetes affects over 300 million people, causing severe complications and premature death, yet the underlying molecular mechanisms are largely unknown. Pancreatic islet dysfunction is central in type 2 diabetes pathogenesis, and understanding islet genome regulation could therefore provide valuable mechanistic insights. We have now mapped and examined the function of human islet cis-regulatory networks. We identify genomic sequences that are targeted by islet transcription factors to drive islet-specific gene activity and show that most such sequences reside in clusters of enhancers that form physical three-dimensional chromatin domains. We find that sequence variants associated with type 2 diabetes and fasting glycemia are enriched in these clustered islet enhancers and identify trait-associated variants that disrupt DNA binding and islet enhancer activity. Our studies illustrate how islet transcription factors interact functionally with the epigenome and provide systematic evidence that the dysregulation of islet enhancers is relevant to the mechanisms underlying type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Cromatina/genética , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Formaldeído , Estudo de Associação Genômica Ampla , Humanos , Dados de Sequência Molecular , Análise de Sequência de RNA , Fatores de Transcrição/genética , Navegador
12.
Nat Genet ; 46(1): 61-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24212882

RESUMO

The contribution of cis-regulatory mutations to human disease remains poorly understood. Whole-genome sequencing can identify all noncoding variants, yet the discrimination of causal regulatory mutations represents a formidable challenge. We used epigenomic annotation in human embryonic stem cell (hESC)-derived pancreatic progenitor cells to guide the interpretation of whole-genome sequences from individuals with isolated pancreatic agenesis. This analysis uncovered six different recessive mutations in a previously uncharacterized ~400-bp sequence located 25 kb downstream of PTF1A (encoding pancreas-specific transcription factor 1a) in ten families with pancreatic agenesis. We show that this region acts as a developmental enhancer of PTF1A and that the mutations abolish enhancer activity. These mutations are the most common cause of isolated pancreatic agenesis. Integrating genome sequencing and epigenomic annotation in a disease-relevant cell type can thus uncover new noncoding elements underlying human development and disease.


Assuntos
Elementos Facilitadores Genéticos/genética , Mutação , Pâncreas/anormalidades , Pancreatopatias/congênito , Fatores de Transcrição/genética , Cromossomos Humanos Par 10 , Células-Tronco Embrionárias/fisiologia , Epigenômica/métodos , Feminino , Genes Recessivos , Humanos , Masculino , Pancreatopatias/genética , Linhagem
13.
J Mater Sci Mater Med ; 24(1): 189-98, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23080375

RESUMO

Cellular microarrays present a promising tool for multiplex evaluation of the signalling effect of substrate-immobilized factors on cellular differentiation. In this paper, we compare the early myoblast-to-osteoblast cell commitment steps in response to a growth factor stimulus using standard well plate differentiation assays or cellular microarrays. Our results show that restraints on the cell culture size, inherent to cellular microarrays, impair the differentiation outcome. Also, while cells growing on spots with immobilised BMP-2 are early biased towards the osteoblast fate, longer periods of cell culturing in the microarrays result in cell proliferation and blockage of osteoblast differentiation. The results presented here raise concerns about the efficiency of cell differentiation when the cell culture dimensions are reduced to a simplified microspot environment. Also, these results suggest that further efforts should be devoted to increasing the complexity of the microspots composition, aiming to replace signalling cues missing in this system.


Assuntos
Diferenciação Celular , Microambiente Celular , Animais , Proteína Morfogenética Óssea 2/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Camundongos , Análise Serial de Proteínas
14.
Methods Mol Biol ; 791: 287-96, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21913087

RESUMO

Noncoding regulatory genomic elements are central for cellular function, differentiation, and disease, but remain poorly characterized. FAIRE (formaldehyde-assisted isolation of regulatory elements) has emerged as a simple method to identify and analyze active regulatory sequences based on their decreased nucleosomal content. More recently FAIRE was combined with high-throughput sequencing (FAIRE-seq) to locate tissue-specific regulatory elements at a genome scale in purified human pancreatic islets. Here we describe the implementation of the FAIRE method in human pancreatic islet cells.


Assuntos
Cromatina/genética , Formaldeído/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Linhagem Celular , Fracionamento Químico , Clorofórmio/química , DNA/genética , DNA/isolamento & purificação , Humanos , Ilhotas Pancreáticas/citologia , Fenol/química , Sonicação , Técnicas de Cultura de Tecidos , Fixação de Tecidos
15.
J Biomed Mater Res A ; 98(2): 245-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21626656

RESUMO

Cellular microarray developments and its applications are the next step after DNA and protein microarrays. The choice of the surface chemistry of the substrates used for the implementation of this technique, that must favor proper protein immobilization while avoiding cell adhesion on the nonspotted areas, presents a complex challenge. This is a key issue since usually the best nonfouling surfaces are also the ones that retain immobilized the smallest amounts of printed protein. To quantitatively assess the amount of protein immobilization, in this study several combinations of fluorescently labeled fibronectin (Fn*) and streptavidin (SA*) were microspotted, with and without glycerol addition in the printing buffer, on several substrates suitable for cellular microarrays. The substrates assayed included chemically activated surfaces as well as Poly ethylene oxide (PEO) films that are nonfouling in solution but accept adhesion of proteins in dry conditions. The results showed that the spotted Fn* was retained by all the surfaces, although the PEO surface did show smaller amounts of immobilization. The SA*, on the other hand, was only retained by the chemically activated surfaces. The inclusion of glycerol in the printing buffer significantly reduced the immobilization of both proteins. The results presented in this article provide quantitative evidence of the convenience of using a chemically activated surface to immobilize proteins relevant for cellular microarray applications, particularly when ECM proteins are cospotted with smaller factors which are more difficult to be retained by the surfaces.


Assuntos
Fibronectinas/análise , Proteínas Imobilizadas/análise , Mioblastos/metabolismo , Estreptavidina/análise , Análise Serial de Tecidos/métodos , Animais , Calibragem , Proliferação de Células , Sobrevivência Celular , Fluorescência , Humanos , Camundongos , Peso Molecular , Mioblastos/citologia
16.
J Mater Sci Mater Med ; 20(7): 1525-33, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19301106

RESUMO

Lately there has been an increasing interest in the development of tools that enable the high throughput analysis of combinations of surface-immobilized signaling factors and which examine their effect on stem cell biology and differentiation. These surface-immobilized factors function as artificial microenvironments that can be ordered in a microarray format. These microarrays could be useful for applications such as the study of stem cell biology to get a deeper understanding of their differentiation process. Here, the evaluation of several key process parameters affecting the cellular microarray fabrication is reported in terms of its effects on the mesenchymal stem cell culture time on these microarrays. Substrate and protein solution requirements, passivation strategies and cell culture conditions are investigated. The results described in this article serve as a basis for the future development of cellular microarrays aiming to provide a deeper understanding of the stem cell differentiation process.


Assuntos
Técnicas de Cultura de Células/instrumentação , Citometria de Fluxo/instrumentação , Análise em Microsséries/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Proteoma/análise , Células-Tronco/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Desenho de Equipamento , Citometria de Fluxo/métodos , Humanos , Análise em Microsséries/métodos , Técnicas Analíticas Microfluídicas/métodos , Células-Tronco/citologia , Avaliação da Tecnologia Biomédica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...