Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Dev Biol ; 417(1): 50-62, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27364467

RESUMO

The mechanisms that modulate and limit the signaling output of adult stem cell niches remain poorly understood. To gain further insights into how these microenvironments are regulated in vivo, we performed a candidate gene screen designed to identify factors that restrict BMP signal production to the cap cells that comprise the germline stem cell (GSC) niche of Drosophila ovaries. Through these efforts, we found that disruption of Wnt4 and components of the canonical Wnt pathway results in a complex germ cell phenotype marked by an expansion of GSC-like cells, pre-cystoblasts and cystoblasts in young females. This phenotype correlates with an increase of decapentaplegic (dpp) mRNA levels within escort cells and varying levels of BMP responsiveness in the germline. Further genetic experiments show that Wnt4, which exhibits graded expression in somatic cells of germaria, activates the Wnt pathway in posteriorly positioned escort cells. The activation of the Wnt pathway appears to be limited by the BMP pathway itself, as loss of Mad in escort cells results in the expansion of Wnt pathway activation. Wnt pathway activity changes within germaria during the course of aging, coincident with changes in dpp production. These data suggest that mutual antagonism between the BMP and Wnt pathways in somatic cells helps to regulate germ cell differentiation.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Células Germinativas/citologia , Glicoproteínas/metabolismo , Ovário/embriologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética , Envelhecimento , Animais , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Feminino , Fenótipo , RNA Mensageiro/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética
2.
3.
PLoS Genet ; 10(3): e1004200, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24625679

RESUMO

Specialized microenvironments called niches regulate tissue homeostasis by controlling the balance between stem cell self-renewal and the differentiation of stem cell daughters. However the mechanisms that govern the formation, size and signaling of in vivo niches remain poorly understood. Loss of the highly conserved histone demethylase Lsd1 in Drosophila escort cells results in increased BMP signaling outside the cap cell niche and an expanded germline stem cell (GSC) phenotype. Here we present evidence that loss of Lsd1 also results in gradual changes in escort cell morphology and their eventual death. To better characterize the function of Lsd1 in different cell populations within the ovary, we performed Chromatin immunoprecipitation coupled with massive parallel sequencing (ChIP-seq). This analysis shows that Lsd1 associates with a surprisingly limited number of sites in escort cells and fewer, and often, different sites in cap cells. These findings indicate that Lsd1 exhibits highly selective binding that depends greatly on specific cellular contexts. Lsd1 does not directly target the dpp locus in escort cells. Instead, Lsd1 regulates engrailed expression and disruption of engrailed and its putative downstream target hedgehog suppress the Lsd1 mutant phenotype. Interestingly, over-expression of engrailed, but not hedgehog, results in an expansion of GSC cells, marked by the expansion of BMP signaling. Knockdown of other potential direct Lsd1 target genes, not obviously linked to BMP signaling, also partially suppresses the Lsd1 mutant phenotype. These results suggest that Lsd1 restricts the number of GSC-like cells by regulating a diverse group of genes and provide further evidence that escort cell function must be carefully controlled during development and adulthood to ensure proper germline differentiation.


Assuntos
Diferenciação Celular/genética , Proteínas de Drosophila/genética , Oxirredutases N-Desmetilantes/genética , Transdução de Sinais/genética , Células-Tronco/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células Germinativas/metabolismo , Histonas/metabolismo , Humanos , Oxirredutases N-Desmetilantes/metabolismo , Nicho de Células-Tronco/genética
4.
Proc Natl Acad Sci U S A ; 108(17): 7064-9, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482791

RESUMO

Specialized microenvironments called niches keep stem cells in an undifferentiated and self-renewing state. Dedicated stromal cells form niches by producing a variety of factors that act directly on stem cells. The size and signaling output of niches must be finely tuned to ensure proper tissue homeostasis. Although advances have been made in identifying factors that promote niche cell fate, the mechanisms that restrict niche cell formation during development and limit niche signaling output in adults remain poorly understood. Here, we show that the histone lysine-specific demethylase 1 (Lsd1) regulates the size of the germline stem cell (GSC) niche in Drosophila ovaries. GSC maintenance depends on bone morphogenetic protein (BMP) signals produced by a small cluster of cap cells located at the anterior tip of the germarium. Lsd1 null mutant ovaries carry small germline tumors containing an expanded number of GSC-like cells with round fusomes that display ectopic BMP signal responsiveness away from the normal niche. Clonal analysis and cell type-specific rescue experiments demonstrate that Lsd1 functions within the escort cells (ECs) that reside immediately adjacent to cap cells and prevents them from ectopically producing niche-specific signals. Temporally restricted gene knockdown experiments suggest that Lsd1 functions both during development, to specify EC fate, and in adulthood, to prevent ECs from forming ectopic niches independent of changes in cell fate. Further analysis shows that Lsd1 functions to repress decapentaplegic (dpp) expression in adult germaria. The role of Lsd1 in regulating niche-specific signals may have important implications for understanding how disruption of its mammalian homolog contributes to cancer and metastasis.


Assuntos
Proteínas de Drosophila/metabolismo , Células-Tronco Neoplásicas/metabolismo , Ovário/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Animais , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Mutação , Células-Tronco Neoplásicas/patologia , Ovário/embriologia , Ovário/patologia , Oxirredutases N-Desmetilantes/genética , Transdução de Sinais/genética
5.
Artigo em Inglês | MEDLINE | ID: mdl-37804247

RESUMO

The geroscience hypothesis suggests that addressing the fundamental mechanisms driving aging biology will prevent or mitigate the onset of multiple chronic diseases, for which the largest risk factor is advanced age. Research that investigates the root causes of aging is therefore of critical importance given the rising healthcare burden attributable to age-related diseases. The third annual Midwest Aging Consortium symposium was convened as a showcase of such research performed by investigators from institutions across the Midwestern United States. This report summarizes the work presented during a virtual conference across topics in aging biology, including immune function in the lung-particularly timely given the Corona Virus Immune Disease-2019 pandemic-along with the role of metabolism and nutrient-regulated pathways in cellular function with age, the influence of senescence on stem cell function and inflammation, and our evolving understanding of the mechanisms underlying observation of sex dimorphism in aging-related outcomes. The symposium focused on early-stage and emerging investigators, while including keynote presentations from leaders in the biology of aging field, highlighting the diversity and strength of aging research in the Midwest.


Assuntos
Envelhecimento , Múltiplas Afecções Crônicas , Humanos , Envelhecimento/fisiologia , Inflamação , Pulmão , Gerociência
6.
Elife ; 112022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36583937

RESUMO

The quiescent muscle stem cell (QSC) pool is heterogeneous and generally characterized by the presence and levels of intrinsic myogenic transcription factors. Whether extrinsic factors maintain the diversity of states across the QSC pool remains unknown. The muscle fiber is a multinucleated syncytium that serves as a niche to QSCs, raising the possibility that the muscle fiber regulates the diversity of states across the QSC pool. Here, we show that the muscle fiber maintains a continuum of quiescent states, through a gradient of Notch ligand, Dll4, produced by the fiber and captured by QSCs. The abundance of Dll4 captured by the QSC correlates with the protein levels of the stem cell (SC) identity marker, Pax7. Niche-specific loss of Dll4 decreases QSC diversity and shifts the continuum to cell states that are biased toward more proliferative and committed fates. We reveal that fiber-derived Mindbomb1 (Mib1), an E3 ubiquitin ligase activates Dll4 and controls the heterogeneous levels of Dll4. In response to injury, with a Dll4-replenished niche, the normal continuum and diversity of the SC pool is restored, demonstrating bidirectionality within the SC continuum. Our data show that a post-translational mechanism controls heterogeneity of Notch ligands in a multinucleated niche cell to maintain a continuum of metastable states within the SC pool during tissue homeostasis.


Assuntos
Células Satélites de Músculo Esquelético , Transdução de Sinais , Fibras Musculares Esqueléticas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Divisão Celular , Células-Tronco/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Nicho de Células-Tronco
7.
Nat Biotechnol ; 24(11): 1392-401, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17053790

RESUMO

Of paramount importance for the development of cell therapies to treat diabetes is the production of sufficient numbers of pancreatic endocrine cells that function similarly to primary islets. We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor--en route to cells that express endocrine hormones. The hES cell-derived insulin-expressing cells have an insulin content approaching that of adult islets. Similar to fetal beta-cells, they release C-peptide in response to multiple secretory stimuli, but only minimally to glucose. Production of these hES cell-derived endocrine cells may represent a critical step in the development of a renewable source of cells for diabetes cell therapy.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células Enteroendócrinas/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Hormônios Pancreáticos/biossíntese , Hormônios Peptídicos/biossíntese , Células Cultivadas , Grelina , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Pâncreas/citologia , Hormônios Pancreáticos/isolamento & purificação
8.
Cell Stem Cell ; 25(5): 654-665.e4, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31495781

RESUMO

Satellite cells (SCs) reside in a dormant state during tissue homeostasis. The specific paracrine agents and niche cells that maintain SC quiescence remain unknown. We find that Wnt4 produced by the muscle fiber maintains SC quiescence through RhoA. Using cell-specific inducible genetics, we find that a Wnt4-Rho signaling axis constrains SC numbers and activation during tissue homeostasis in adult mice. Wnt4 activates Rho in quiescent SCs to maintain mechanical strain, restrict movement in the niche, and repress YAP. The induction of YAP upon disruption of RhoA is essential for SC activation under homeostasis. In the context of injury, the loss of Wnt4 from the niche accelerates SC activation and muscle repair, whereas overexpression of Wnt4 transitions SCs into a deeper state of quiescence and delays muscle repair. In conclusion, the SC pool undergoes dynamic transitions during early activation with changes in mechano-properties and cytoskeleton signaling preceding cell-cycle entry.


Assuntos
Proliferação de Células/genética , Fibras Musculares Esqueléticas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Proteína Wnt4/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/fisiologia , Citoesqueleto/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Força Atômica , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Regeneração/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais/genética , Nicho de Células-Tronco/genética , Proteína Wnt4/genética , Proteínas de Sinalização YAP
9.
Nat Biotechnol ; 23(12): 1534-41, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16258519

RESUMO

The potential of human embryonic stem (hES) cells to differentiate into cell types of a variety of organs has generated much excitement over the possible use of hES cells in therapeutic applications. Of great interest are organs derived from definitive endoderm, such as the pancreas. We have focused on directing hES cells to the definitive endoderm lineage as this step is a prerequisite for efficient differentiation to mature endoderm derivatives. Differentiation of hES cells in the presence of activin A and low serum produced cultures consisting of up to 80% definitive endoderm cells. This population was further enriched to near homogeneity using the cell-surface receptor CXCR4. The process of definitive endoderm formation in differentiating hES cell cultures includes an apparent epithelial-to-mesenchymal transition and a dynamic gene expression profile that are reminiscent of vertebrate gastrulation. These findings may facilitate the use of hES cells for therapeutic purposes and as in vitro models of development.


Assuntos
Técnicas de Cultura de Células/métodos , Endoderma/citologia , Endoderma/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Camundongos
10.
Mol Cell Biol ; 23(2): 482-92, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12509448

RESUMO

The chimeric fusion gene EWS/FLI-1 is detected in most cases of Ewing's sarcoma (ES), the second most common malignant bone tumor of childhood. Although 80% of ES tumors develop in skeletal sites, the remainder can arise in almost any soft tissue location. The lineage of the cell developing the EWS/FLI-1 gene fusion has not been fully characterized but is generally considered to be of either mesenchymal or neural crest origin. To study this oncogene in a conceptually relevant target cell, EWS/FLI-1 was introduced into the murine cell line C2C12, a myoblast cell line capable of differentiation into muscle, bone, or fat. In this cellular context, EWS/FLI-1 profoundly inhibited the myogenic differentiation program. The block in C2C12 myogenic differentiation required the nuclear localization and DNA-binding functions of EWS/FLI-1 and was mediated by transcriptional and posttranscriptional suppression of the myogenic transcription factors MyoD and myogenin. Interestingly, C2C12-EWS/FLI-1 cells constitutively expressed alkaline phosphatase, a bone lineage marker, and were alkaline phosphatase positive by histochemistry but showed no other evidence of bone lineage commitment. Consistent with recent findings in human ES tumor cell lines, C2C12-EWS/FLI-1 cells constitutively expressed cyclin D1 and demonstrated decreased expression of the cell cycle regulator p21(cip1), even under differentiation conditions and at confluent density. This C2C12-EWS/FLI-1 cell model may assist in the identification of novel differentially expressed genes relevant to ES and provide further insight into the cell(s) of origin developing ES-associated genetic fusions.


Assuntos
Proteínas de Fusão Oncogênica/fisiologia , Sarcoma de Ewing/metabolismo , Fatores de Transcrição/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Western Blotting , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Citometria de Fluxo , Vetores Genéticos , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Proteína MyoD/metabolismo , Miogenina/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Oncogenes/genética , Plasmídeos/metabolismo , Proteína Proto-Oncogênica c-fli-1 , Proteína EWS de Ligação a RNA , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção
11.
Cancer Res ; 65(19): 8698-705, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16204038

RESUMO

Ewing sarcoma is the second most common malignant pediatric bone tumor. Over 80% of Ewing sarcoma contain the oncogene EWS/FLI-1, which encodes the EWS/FLI-1 oncoprotein, a hybrid transcription factor comprised of NH2-terminal sequences from the RNA-binding protein EWS and the DNA-binding and COOH-terminal regions of the Ets transcription factor FLI-1. Although numerous genes are dysregulated by EWS/FLI-1, advances in Ewing sarcoma cancer biology have been hindered by the lack of an animal model because of EWS/FLI-1-mediated cytotoxicity. In this study, we have developed conditions for the isolation and propagation of murine primary bone-derived cells (mPBDC) that stably express EWS/FLI-1. Early-passage EWS/FLI-1 mPBDCs were immortalized in culture but inefficient at tumor induction, whereas later-passage cells formed sarcomatous tumors in immunocompetent syngeneic mice. Murine EWS/FLI-1 tumors contained morphologically primitive cells that lacked definitive lineage markers. Molecular characterization of murine EWS/FLI-1 tumors revealed that some but not all had acquired a novel, clonal in-frame p53 mutation associated with a constitutive loss of p21 expression. Despite indications that secondary events facilitated EWS/FLI-1 mPBDC tumorigenesis, cells remained highly dependent on EWS/FLI-1 for efficient transformation in clonogenic assays. This Ewing sarcoma animal model will be a useful tool for dissecting the molecular pathogenesis of Ewing sarcoma and provides rationale for the broader use of organ-specific progenitor cell populations for the study of human sarcoma.


Assuntos
Neoplasias Ósseas/metabolismo , Proteínas de Fusão Oncogênica/biossíntese , Proteína Proto-Oncogênica c-fli-1/biossíntese , Proteína EWS de Ligação a RNA/biossíntese , Sarcoma de Ewing/metabolismo , Sequência de Aminoácidos , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Ciclo Celular/fisiologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas de Fusão Oncogênica/genética , Proteína Proto-Oncogênica c-fli-1/genética , Proteína EWS de Ligação a RNA/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia
12.
Sci Rep ; 7(1): 5240, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701701

RESUMO

Jumonji (JmjC) domain proteins influence gene expression and chromatin organization by way of histone demethylation, which provides a means to regulate the activity of genes across the genome. JmjC proteins have been associated with many human diseases including various cancers, developmental and neurological disorders, however, the shared biology and possible common contribution to organismal development and tissue homeostasis of all JmjC proteins remains unclear. Here, we systematically tested the function of all 13 Drosophila JmjC genes. Generation of molecularly defined null mutants revealed that loss of 8 out of 13 JmjC genes modify position effect variegation (PEV) phenotypes, consistent with their ascribed role in regulating chromatin organization. However, most JmjC genes do not critically regulate development, as 10 members are viable and fertile with no obvious developmental defects. Rather, we find that different JmjC mutants specifically alter the phenotypic outcomes in various sensitized genetic backgrounds. Our data demonstrate that, rather than controlling essential gene expression programs, Drosophila JmjC proteins generally act to "fine-tune" different biological processes.


Assuntos
Animais Geneticamente Modificados/genética , Cromatina/química , Metilação de DNA , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Animais Geneticamente Modificados/crescimento & desenvolvimento , Animais Geneticamente Modificados/metabolismo , Cromatina/genética , Cromatina/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Redes Reguladoras de Genes , Histonas/metabolismo , Masculino , Transdução de Sinais
13.
Cell Stem Cell ; 18(1): 5-7, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26748748

RESUMO

Quiescence is highly regulated to preserve stem cell function. In this issue of Cell Stem Cell, Zismanov et al. (2016) show that P-eIF2α, a translational initiation factor, reinforces the quiescent state of muscle stem cells by safeguarding against cell-cycle entry and lineage progression.


Assuntos
Diferenciação Celular , Células Satélites de Músculo Esquelético , Ciclo Celular , Humanos , Células-Tronco/citologia
14.
Stem Cell Res Ther ; 2(6): 45, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22117545

RESUMO

Specialized microenvironments called niches help maintain stem cells in an undifferentiated and self-renewing state. The existence of niches has long been predicted from mammalian studies, but identifying stem cells in their native environments in vivo has remained a challenge in most vertebrates. Many of the mechanistic insights into how niches regulate stem cell maintenance have been obtained using invertebrate models such as Drosophila. Here, we focus on the Drosophila ovarian germline stem cell niche and review recent studies that have begun to reveal how intricate crosstalk between various signaling pathways regulates stem cell maintenance, how the extracellular matrix modulates the signaling output of the niche and how epigenetic programming influences cell development and function both inside and outside the niche to ensure proper tissue homeostasis. These insights will probably inform the study of mammalian niches and how their malfunction contributes to human disease.


Assuntos
Ovário/citologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/fisiologia , Epigenômica , Matriz Extracelular/metabolismo , Feminino , Insulina/metabolismo , Ovário/metabolismo , Transdução de Sinais , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
15.
Nat Biotechnol ; 26(4): 443-52, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18288110

RESUMO

Development of a cell therapy for diabetes would be greatly aided by a renewable supply of human beta-cells. Here we show that pancreatic endoderm derived from human embryonic stem (hES) cells efficiently generates glucose-responsive endocrine cells after implantation into mice. Upon glucose stimulation of the implanted mice, human insulin and C-peptide are detected in sera at levels similar to those of mice transplanted with approximately 3,000 human islets. Moreover, the insulin-expressing cells generated after engraftment exhibit many properties of functional beta-cells, including expression of critical beta-cell transcription factors, appropriate processing of proinsulin and the presence of mature endocrine secretory granules. Finally, in a test of therapeutic potential, we demonstrate that implantation of hES cell-derived pancreatic endoderm protects against streptozotocin-induced hyperglycemia. Together, these data provide definitive evidence that hES cells are competent to generate glucose-responsive, insulin-secreting cells.


Assuntos
Técnicas de Cultura de Células/tendências , Células-Tronco Embrionárias/citologia , Glucose/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Engenharia Tecidual/tendências , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Endoderma/citologia , Endoderma/metabolismo , Humanos , Células Secretoras de Insulina/transplante , Camundongos , Pâncreas Artificial/tendências
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa