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.
Gene Expr Patterns ; 48: 119319, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37148985

RESUMO

Heterogeneous nuclear ribonucleoprotein L (hnRNPL) is a conserved RNA binding protein (RBP) that plays an important role in the alternative splicing of gene transcripts, and thus in the generation of specific protein isoforms. Global deficiency in hnRNPL in mice results in preimplantation embryonic lethality at embryonic day (E) 3.5. To begin to understand the contribution of hnRNPL-regulated pathways in the normal development of the embryo and placenta, we determined hnRNPL expression profile and subcellular localization throughout development. Proteome and Western blot analyses were employed to determine hnRNPL abundance between E3.5 and E17.5. Histological analyses supported that the embryo and implantation site display distinct hnRNPL localization patterns. In the fully developed mouse placenta, nuclear hnRNPL was observed broadly in trophoblasts, whereas within the implantation site a discrete subset of cells showed hnRNPL outside the nucleus. In the first-trimester human placenta, hnRNPL was detected in the undifferentiated cytotrophoblasts, suggesting a role for this factor in trophoblast progenitors. Parallel in vitro studies utilizing Htr8 and Jeg3 cell lines confirmed expression of hnRNPL in cellular models of human trophoblasts. These studies [support] coordinated regulation of hnRNPL during the normal developmental program in the mammalian embryo and placenta.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L , Placenta , Animais , Feminino , Humanos , Camundongos , Gravidez , Linhagem Celular Tumoral , Embrião de Mamíferos , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo
2.
Exp Neurol ; 351: 113986, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35065053

RESUMO

Microglia are resident immune cells in the central nervous system (CNS), which, in a healthy state, promote CNS homeostasis and respond to CNS injury. In contrast, microglia are also implicated in pathological conditions where they may contribute to neural damage. Primitive microglia arise from extraembryonic progenitors in the yolk sac (YS). The extraembryonic origins of primitive microglia are distinct from other tissue macrophages. The YS is the first site of hematopoiesis in development. Uniquely, microglial pregenital cells in the mouse derive from an early myeloid branch of the hematopoietic lineage in the YS. Microglia are critical in several key stages of physiological brain development, including embryonic vasculogenesis, immunosurveillance, and neurogenesis. Abnormal microglial function has been linked to neurodevelopmental and neurodegenerative diseases, although mechanistic roles in disease etiology remain incompletely understood. Knowledge of species-specific differences between human, murine and other animal models is also critical to understanding translational relevance to human health and disease as biomedical understanding of the importance of primitive microglia advances. This significance drives the importance of understanding, comparatively, the extraembryonic origins and developmental mechanisms whereby human primitive microglia differentiate and migrate to inform translational research. A better understanding of the molecular drivers may lead to biomarkers and/or preventative or therapeutic measures for neonatal brain development and neurodegenerative diseases. Herein, the role of microglia in neonatal brain development is discussed, current understandings of the developmental origins of microglia are described, the ontogeny and phylogeny of microglia, and implications of in vitro microglia-like cell differentiation, with a specific interest on neurodegenerative diseases, are reviewed. Together, these emphasize the importance of leveraging the extraembryonic origins of microglia to not only better understand neurodevelopment and neurodegenerative diseases, but also to develop protective measures that are specific to human microglia.


Assuntos
Macrófagos , Microglia , Animais , Encéfalo , Hematopoese/fisiologia , Camundongos , Saco Vitelino/fisiologia
3.
Curr Stem Cell Rep ; 7(4): 185-193, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34697582

RESUMO

Purpose of Review: The placenta is a transient organ that forms de novo and serves a critical role in supporting fetal growth and development. Placental oxygen, nutrients, and waste are transported through processes that depend on vascular structure and cell type-specific expression and localization of membrane transporters. Understanding how the placenta develops holds great significance for maternal-fetal medicine. The purpose of this review is to examine current information regarding placental progenitor populations. Recent Findings: Recent advancements in single-cell RNA sequencing (scRNA-seq) provide unprecedented depth for the investigation of cell type-specific gene expression patterns in the placenta. Thus far, several mouse placenta scRNA-seq studies have been conducted which produced and analyzed transcriptomes of placental progenitors and cells of the fully developed placenta between embryonic day (E) 7.0 and E12.5. Together with human placenta scRNA-seq data which, in part, has been produced through coordinated research campaigns in the scientific community to understand the potential for SARS-CoV-2 infection, these mammalian studies lend fundamental insight into the cellular and molecular composition of hemochorial placentae found in both mouse and human. Summary: Single-cell placenta research has advanced understanding of tissue-resident stem cells and molecules that are poised to support maternal-fetal communication and nutrient transport. Herein, we provide context for these recent findings by reviewing placental anatomy and cell populations, and discuss recent scRNA-seq mouse placenta findings. Further research is needed to evaluate the utility of placental stem cells in the development of new therapeutic approaches for the treatment of wound healing and disease.

4.
Exp Dermatol ; 30(8): 1065-1072, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34114688

RESUMO

Diabetic foot ulcers (DFUs), a prevalent complication of diabetes, constitute a major medical challenge with a critical need for development of cell-based therapies. We previously generated induced pluripotent stem cells (iPSCs) from dermal fibroblasts derived from the DFU patients, location-matched skin of diabetic patients and normal healthy donors and re-differentiated them into fibroblasts. To assess the epigenetic microRNA (miR) regulated changes triggered by cellular reprogramming, we performed miRs expression profiling. We found let-7c, miR-26b-5p, -29c-3p, -148a-3p, -196a-5p, -199b-5p and -374a-5p suppressed in iPSC-derived fibroblasts in vitro and in 3D dermis-like self-assembly tissue, whereas their corresponding targets involved in cellular migration were upregulated. Moreover, targets involved in organization of extracellular matrix were induced after fibroblast reprogramming. PLAT gene, the crucial fibrinolysis factor, was upregulated in iPSC-derived fibroblasts and was confirmed as a direct target of miR-196a-5p. miR-197-3p and miR-331-3p were found upregulated specifically in iPSC-derived diabetic fibroblasts, while their targets CAV1 and CDKN3 were suppressed. CAV1, an important negative regulator of wound healing, was confirmed as a direct miR-197-3p target. Together, our findings demonstrate that iPSC reprogramming is an effective approach for erasing the diabetic non-healing miR-mediated epigenetic signature and promoting a pro-healing cellular phenotype.


Assuntos
Reprogramação Celular/genética , Pé Diabético/genética , Epigênese Genética , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/genética , Cicatrização/genética , Movimento Celular/genética , Humanos , Regulação para Cima
5.
Tissue Eng Part C Methods ; 27(2): 49-58, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33280487

RESUMO

A major challenge in the management of patients suffering from diabetes is the risk of developing nonhealing foot ulcers. Most in vitro methods to screen drugs for wound healing therapies rely on conventional 2D cell cultures that do not closely mimic the complexity of the diabetic wound environment. In addition, while three-dimensional (3D) skin tissue models of human skin exist, they have not previously been adapted to incorporate patient-derived macrophages to model inflammation from these wounds. In this study, we present a 3D human skin equivalent (HSE) model incorporating blood-derived monocytes and primary fibroblasts isolated from patients with diabetic foot ulcers (DFUs). We demonstrate that the monocytes differentiate into macrophages when incorporated into HSEs and secrete a cytokine profile indicative of the proinflammatory M1 phenotype seen in DFUs. We also show how the interaction between fibroblasts and macrophages in the HSE can guide macrophage polarization. Our findings take us a step closer to creating a human, 3D skin-like tissue model that can be applied to evaluate the response of candidate compounds needed for potential new foot ulcer therapies in a more complex tissue environment that contributes to diabetic wounds. Impact statement This study is the first to incorporate disease-specific, diabetic macrophages into a three-dimensional (3D) model of human skin. We show how to fabricate skin that incorporates macrophages with disease-specific fibroblasts to guide macrophage polarization. We also show that monocytes from diabetic patients can differentiate into macrophages directly in this skin disease model, and that they secrete a cytokine profile mimicking the proinflammatory M1 phenotype seen in diabetic foot ulcers. The data presented here indicate that this 3D skin disease model can be used to study macrophage-related inflammation in diabetes and as a drug testing tool to evaluate new treatments for the disease.


Assuntos
Diabetes Mellitus , Pé Diabético , Fibroblastos , Humanos , Macrófagos , Pele , Cicatrização
6.
Adv Healthc Mater ; 9(16): e2000307, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32597577

RESUMO

Diabetic foot ulcers (DFUs) are chronic wounds, with 20% of cases resulting in amputation, despite intervention. A recently approved tissue engineering product-a cell-free collagen-glycosaminoglycan (GAG) scaffold-demonstrates 50% success, motivating its functionalization with extracellular matrix (ECM). Induced pluripotent stem cell (iPSC) technology reprograms somatic cells into an embryonic-like state. Recent findings describe how iPSCs-derived fibroblasts ("post-iPSF") are proangiogenic, produce more ECM than their somatic precursors ("pre-iPSF"), and their ECM has characteristics of foetal ECM (a wound regeneration advantage, as fetuses heal scar-free). ECM production is 45% higher from post-iPSF and has favorable components (e.g., Collagen I and III, and fibronectin). Herein, a freeze-dried scaffold using ECM grown by post-iPSF cells (Post-iPSF Coll) is developed and tested vs precursors ECM-activated scaffolds (Pre-iPSF Coll). When seeded with healthy or DFU fibroblasts, both ECM-derived scaffolds have more diverse ECM and more robust immune responses to cues. Post-iPSF-Coll had higher GAG, higher cell content, higher Vascular Endothelial Growth Factor (VEGF) in DFUs, and higher Interleukin-1-receptor antagonist (IL-1ra) vs. pre-iPSF Coll. This work constitutes the first step in exploiting ECM from iPSF for tissue engineering scaffolds.


Assuntos
Diabetes Mellitus , Células-Tronco Pluripotentes Induzidas , Matriz Extracelular , Fibroblastos , Humanos , Engenharia Tecidual , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular , Cicatrização
7.
FASEB J ; 33(1): 1262-1277, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30088952

RESUMO

Diabetic foot ulcers (DFUs) are a major complication of diabetes, and there is a critical need to develop novel cell- and tissue-based therapies to treat these chronic wounds. Induced pluripotent stem cells (iPSCs) offer a replenishing source of allogeneic and autologous cell types that may be beneficial to improve DFU wound-healing outcomes. However, the biologic potential of iPSC-derived cells to treat DFUs has not, to our knowledge, been investigated. Toward that goal, we have performed detailed characterization of iPSC-derived fibroblasts from both diabetic and nondiabetic patients. Significantly, gene array and functional analyses reveal that iPSC-derived fibroblasts from both patients with and those without diabetes are more similar to each other than were the primary cells from which they were derived. iPSC-derived fibroblasts showed improved migratory properties in 2-dimensional culture. iPSC-derived fibroblasts from DFUs displayed a unique biochemical composition and morphology when grown as 3-dimensional (3D), self-assembled extracellular matrix tissues, which were distinct from tissues fabricated using the parental DFU fibroblasts from which they were reprogrammed. In vivo transplantation of 3D tissues with iPSC-derived fibroblasts showed they persisted in the wound and facilitated diabetic wound closure compared with primary DFU fibroblasts. Taken together, our findings support the potential application of these iPSC-derived fibroblasts and 3D tissues to improve wound healing.-Kashpur, O., Smith, A., Gerami-Naini, B., Maione, A. G., Calabrese, R., Tellechea, A., Theocharidis, G., Liang, L., Pastar, I., Tomic-Canic, M., Mooney, D., Veves, A., Garlick, J. A. Differentiation of diabetic foot ulcer-derived induced pluripotent stem cells reveals distinct cellular and tissue phenotypes.


Assuntos
Diferenciação Celular , Pé Diabético/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Pé Diabético/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos SCID , Fenótipo , Cicatrização/genética
8.
Lab Invest ; 99(4): 514-527, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30568176

RESUMO

Cutaneous fibrosis is a common complication seen in mixed connective tissue diseases. It often occurs as a result of TGF-ß-induced deposition of excessive amounts of collagen in the skin. Lysyl oxidases (LOXs), a family of extracellular matrix (ECM)-modifying enzymes responsible for collagen cross-linking, are known to be increased in dermal fibroblasts from patients with fibrotic diseases, denoting a possible role of LOXs in fibrosis. To directly study this, we have developed two bioengineered, in vitro skin-like models: human skin equivalents (hSEs), and self-assembled stromal tissues (SASs) that contain either normal or systemic sclerosis (SSc; scleroderma) patient-derived fibroblasts. These tissues provide an organ-level structure that could be combined with non-invasive, label-free, multiphoton microscopy (SHG/TPEF) to reveal alterations in the organization and cross-linking levels of collagen fibers during the development of cutaneous fibrosis, which demonstrated increased stromal rigidity and activation of dermal fibroblasts in response to TGF-ß1. Specifically, inhibition of specific LOXs isoforms, LOX and LOXL4, in foreskin fibroblasts (HFFs) resulted in antagonistic effects on TGF-ß1-induced fibrogenic hallmarks in both hSEs and SASs. In addition, a translational relevance of these models was seen as similar antifibrogenic phenotypes were achieved upon knocking down LOXL4 in tissues containing SSc patient-derived-dermal fibroblasts (SScDFs). These findings point to a pivotal role of LOXs in TGF-ß1-induced cutaneous fibrosis through impaired ECM homeostasis in skin-like tissues, and show the value of these tissue platforms in accelerating the discovery of antifibrosis therapeutics.


Assuntos
Fibroblastos/metabolismo , Fibrose/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Aminoácido Oxirredutases/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Fibroblastos/citologia , Humanos , Modelos Biológicos , Fenótipo , Pele/citologia , Pele/metabolismo
9.
Cell Reprogram ; 18(4): 214-23, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27328415

RESUMO

Diabetic foot ulcers (DFUs) are nonhealing chronic wounds that are a serious complication of diabetes. Since induced pluripotent stem cells (iPSCs) may offer a potent source of autologous cells to heal these wounds, we studied if repair-deficient fibroblasts, derived from DFU patients and age- and site-matched control fibroblasts, could be reprogrammed to iPSCs. To establish this, we used Sendai virus to successfully reprogram six primary fibroblast cell lines derived from ulcerated skin of two DFU patients (DFU8, DFU25), nonulcerated foot skin from two diabetic patients (DFF24, DFF9), and healthy foot skin from two nondiabetic patients (NFF12, NFF14). We confirmed reprogramming to a pluripotent state through three independent criteria: immunofluorescent staining for SSEA-4 and TRA-1-81, formation of embryoid bodies with differentiation potential to all three embryonic germ layers in vitro, and formation of teratomas in vivo. All iPSC lines showed normal karyotypes and typical, nonmethylated CpG sites for OCT4 and NANOG. iPSCs derived from DFUs were similar to those derived from site-matched nonulcerated skin from both diabetic and nondiabetic patients. These results have established for the first time that multiple, DFU-derived fibroblast cell lines can be reprogrammed with efficiencies similar to control fibroblasts, thus demonstrating their utility for future regenerative therapy of DFUs.


Assuntos
Diferenciação Celular , Reprogramação Celular , Pé Diabético/patologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Vírus Sendai/genética , Teratoma/patologia , Animais , Células Cultivadas , Pé Diabético/genética , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos SCID , Teratoma/etiologia
10.
Wound Repair Regen ; 24(4): 630-43, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27102877

RESUMO

Current chronic wound treatments often fail to promote healing of diabetic foot ulcers (DFU), leading to amputation and increased patient morbidity. A critical mediator of proper wound healing is the production, assembly, and remodeling of the extracellular matrix (ECM) by fibroblasts. However, little is known about how these processes are altered in fibroblasts within the DFU microenvironment. Thus, we investigated the capacity of multiple, primary DFU-derived fibroblast strains to express, produce, and assemble ECM proteins compared to diabetic patient-derived fibroblasts and healthy donor-derived fibroblasts. Gene expression microarray analysis showed differential expression of ECM and ECM-regulatory genes by DFU-derived fibroblasts which translated to functional differences in a 3D in vitro ECM tissue model. DFU-derived fibroblasts produced thin, fibronectin-rich matrices, and responded abnormally when challenged with transforming growth factor-beta, a key regulator of matrix production during healing. These results provide novel evidence that DFU-derived fibroblasts contribute to the defective matrices of DFUs and chronic wound pathogenesis.


Assuntos
Pé Diabético/patologia , Pé Diabético/fisiopatologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Cicatrização , Colágeno Tipo I/metabolismo , Pé Diabético/metabolismo , Matriz Extracelular/química , Matriz Extracelular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Análise em Microsséries , Neovascularização Fisiológica , Fator de Crescimento Transformador beta/farmacologia
11.
PLoS One ; 9(2): e89546, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586860

RESUMO

The POU5F1 gene codes for the OCT4 transcription factor, which is one of the key regulators of pluripotency. Its transcription, alternative splicing, and alternative translation leading to the synthesis of the active, nuclear localized OCT4A has been described in detail. Much less, however, is known about actively transcribed OCT4 pseudogenes, several of which display high homology to OCT4A and can be expressed and translated into proteins. Using RT-PCR followed by pseudogene-specific restriction digestion, cloning, and sequencing we discriminate between OCT4A and transcripts for pseudogenes 1, 3 and 4. We show that expression of OCT4 and its pseudogenes follows a developmentally-regulated pattern in differentiating hESCs, indicating a tight regulatory relationship between them. We further demonstrate that differentiated human cells from a variety of tissues express exclusively pseudogenes. Expression of OCT4A can, however be triggered in adult differentiated cells by oxygen and FGF2-dependent mechanisms.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Pseudogenes/fisiologia , Adulto , Western Blotting , Proliferação de Células , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Oxigênio/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
BMC Genomics ; 14: 656, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24066673

RESUMO

BACKGROUND: Adult human fibroblasts grown in low oxygen and with FGF2 supplementation have the capacity to tip the healing outcome of skeletal muscle injury - by favoring regeneration response in vivo over scar formation. Here, we compare the transcriptomes of control adult human dermal fibroblasts and induced regeneration-competent (iRC) fibroblasts to identify transcriptional changes that may be related to their regeneration competence. RESULTS: We identified a unique gene-expression profile that characterizes FGF2-induced iRC fibroblast phenotype. Significantly differentially expressed genes due to FGF2 treatment were identified and analyzed to determine overrepresented Gene Ontology terms. Genes belonging to extracellular matrix components, adhesion molecules, matrix remodelling, cytoskeleton, and cytokines were determined to be affected by FGF2 treatment. CONCLUSIONS: Transcriptome analysis comparing control adult human fibroblasts with FGF2-treated fibroblasts identified functional groups of genes that reflect transcriptional changes potentially contributing to their regeneration competence. This comparative transcriptome analysis should contribute new insights into genes that characterize cells with greater regenerative potential.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Regeneração/genética , Transcriptoma/genética , Adulto , Moléculas de Adesão Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Ontologia Genética , Humanos , Receptores de Citocinas/metabolismo , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Cicatrização/genética
13.
Int J Dev Biol ; 54(11-12): 1743-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21136380

RESUMO

The transcription factor NANOG is essential for maintaining pluripotency in embryonic stem cells. We have previously reported the expression of NANOG in adult human fibroblasts; here we present a more thorough investigation into the expression of NANOG in a panel of both differentiated and undifferentiated human cells. We utilize RT-PCR, qRT-PCR, cloning and sequencing, sequence alignment, restriction digestion, immunocytochemistry, Western blotting, and EMSA to investigate expression of NANOG in a variety of somatic, transformed and stem cell phenotypes. RT-PCR and qRT-PCR analysis revealed the presence of NANOG transcripts in all the cell types examined, albeit at magnitudes lower than human embryonic stem cells. Further investigation by single nucleotide polymorphism analysis of expressed transcripts in several cell types detected a NANOG pseudogene, NANOGP8, one of only two NANOG pseudogenes with the potential of encoding a similar size protein to embryonic NANOG (eNANOG). Our analysis demonstrates that although the NANOG protein is detected in nearly all cells examined, expression of the eNANOG and/or NANOGP8 transcript as well as the sub-cellular localization of the protein is cell type-specific. Additionally, smooth muscle cells, which express exclusively NANOGP8, display nuclear localization of NANOG protein, indicating that NANOGP8 is a protein coding gene possibly functioning as a transcription factor. Lastly, all cell types expressing eNANOG and/or NANOGP8 were found to be capable of binding a NANOG consensus sequence in vitro. We conclude that eNANOG is not exclusively expressed in undifferentiated cells and that both eNANOG and NANOGP8 may function as transcription factors in a cell type-specific manner.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/genética , Pseudogenes/genética , Sequência de Aminoácidos , Sequência Consenso/genética , Ensaio de Desvio de Mobilidade Eletroforética , Fibroblastos/classificação , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Células HeLa , Proteínas de Homeodomínio/química , Humanos , Dados de Sequência Molecular , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteína Homeobox Nanog , Polimorfismo de Nucleotídeo Único , Alinhamento de Sequência
14.
Genetics ; 185(1): 211-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20233857

RESUMO

Many plant-associated microbes synthesize the auxin indole-3-acetic acid (IAA), and several IAA biosynthetic pathways have been identified in microbes and plants. Saccharomyces cerevisiae has previously been shown to respond to IAA by inducing pseudohyphal growth. We observed that IAA also induced hyphal growth in the human pathogen Candida albicans and thus may function as a secondary metabolite signal that regulates virulence traits such as hyphal transition in pathogenic fungi. Aldehyde dehydrogenase (Ald) is required for IAA synthesis from a tryptophan (Trp) precursor in Ustilago maydis. Mutant S. cerevisiae with deletions in two ALD genes are unable to convert radiolabeled Trp to IAA, yet produce IAA in the absence of exogenous Trp and at levels higher than wild type. These data suggest that yeast may have multiple pathways for IAA synthesis, one of which is not dependent on Trp.


Assuntos
Ácidos Indolacéticos/metabolismo , Morfogênese , Característica Quantitativa Herdável , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/patogenicidade , Candida albicans/citologia , Candida albicans/efeitos dos fármacos , Diploide , Deleção de Genes , Genes Fúngicos/genética , Testes Genéticos , Homeostase/efeitos dos fármacos , Humanos , Ácidos Indolacéticos/química , Redes e Vias Metabólicas/efeitos dos fármacos , Morfogênese/efeitos dos fármacos , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Triptofano/farmacologia , Virulência/efeitos dos fármacos
15.
Cloning Stem Cells ; 11(3): 417-26, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19622035

RESUMO

Reprogramming of differentiated somatic cells into induced pluripotent stem (iPS) cells has potential for derivation of patient-specific cells for therapy as well as for development of models with which to study disease progression. Derivation of iPS cells from human somatic cells has been achieved by viral transduction of human fibroblasts with early developmental genes. Because forced expression of these genes by viral transduction results in transgene integration with unknown and unpredictable potential mutagenic effects, identification of cell culture conditions that can induce endogenous expression of these genes is desirable. Here we show that primary adult human fibroblasts have basal expression of mRNA for OCT4, SOX2, and NANOG. However, translation of these messages into detectable proteins and their subcellular localization depends on cell culture conditions. Manipulation of oxygen concentration and FGF2 supplementation can modulate expression of some pluripotency related genes at the transcriptional, translational, and cellular localization level. Changing cell culture condition parameters led to expression of REX1, potentiation of expression of LIN28, translation of OCT4, SOX2, and NANOG, and translocation of these transcription factors to the cell nucleus. We also show that culture conditions affect the in vitro lifespan of dermal fibroblasts, nearly doubling the number of population doublings before the cells reach replicative senescence. Our results suggest that it is possible to induce and manipulate endogenous expression of stem cell genes in somatic cells without genetic manipulation, but this short-term induction may not be sufficient for acquisition of true pluripotency. Further investigation of the factors involved in inducing this response could lead to discovery of defined culture conditions capable of altering cell fate in vitro. This would alleviate the need for forced expression by transgenesis, thus eliminating the risk of mutagenic effects due to genetic manipulation.


Assuntos
Desdiferenciação Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Oxigênio/farmacologia , Adulto , Proteínas de Transporte/biossíntese , Técnicas de Cultura de Células , Ciclo-Oxigenase 2/biossíntese , Fibroblastos/metabolismo , Proteínas de Homeodomínio , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Proteína Homeobox Nanog , Proteínas Nucleares/biossíntese , Fator 3 de Transcrição de Octâmero/biossíntese , Proteínas de Ligação a RNA/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...