Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.457
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 32(15-16): 1008-1019, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30042132

RESUMO

Various types of repetitive sequences are dysregulated in cancer. In Ewing sarcoma, the oncogenic fusion protein EWS-FLI1 induces chromatin features typical of active enhancers at GGAA microsatellite repeats, but the function of these sites has not been directly demonstrated. Here, by combining nascent transcription profiling with epigenome editing, we found that a subset of GGAA microsatellite repeats is transcriptionally active in Ewing sarcoma and that silencing individual repeats abolishes local nascent transcription and leads to markedly reduced expression of putative target genes. Epigenome silencing of these repeat sites does not affect gene expression in unrelated cells, can prevent the induction of gene expression by EWS-FLI1, and, in the case of a GGAA repeat that controls SOX2 expression from a distance of 470 kb, is sufficient to impair the growth of Ewing sarcoma xenografts. Using an experimental approach that is broadly applicable to testing different types of repetitive genomic elements, our study directly demonstrates that specific repeat microsatellites can have critical gene regulation functions in cancer and thus represent tumor-specific vulnerabilities that may be exploited to develop new therapies.


Assuntos
Neoplasias Ósseas/genética , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Repetições de Microssatélites , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/genética , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Células Cultivadas , Cromatina/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , RNA não Traduzido/biossíntese , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transcrição Gênica , Células Tumorais Cultivadas , Proteínas de Peixe-Zebra
2.
Arkh Patol ; 86(3): 12-20, 2024.
Artigo em Russo | MEDLINE | ID: mdl-38881001

RESUMO

OBJECTIVE: To study the features of gastric neuroendocrine tumors (NETs) and the diagnostic and prognostic significance of PDX-1 expression in them. MATERIAL AND METHODS: 207 NETs identified in 56 men and 115 women (59 had multiple NETs), and 94 cases of gastric cancer (comparison group) were studied morphologically and immunohistochemically. RESULTS: In more than half of the cases (54.93%), NETs were localized in the body of the stomach; the cardiac and antral parts of the stomach accounted for 8.64% and 11.73%, respectively. NETs of the cardiac region predominated in men, and of the body and antrum - in women. NETs of the cardiac region predominated in men, and of the body and antrum - in women. The vast majority of NETs were highly differentiated (89.20%), of which Grade 1, 2 and 3 were 55.41%, 40.76% and 3.82%, respectively. Neuroendocrine carcinomas (NEC) accounted for 10.80% of all NET cases. NECs were more often localized in the cardiac part of the stomach and accounted for 35.71% of all NETs in the cardiac part. The share of NEC among all NETs of the antrum was 15.79%, of the body of the stomach - only 3.37%. Metastases were found in 17.90% of NETs. Expression of PDX-1 was detected in 44.73% of NETs, 70% of NECs and 74.50% of gastric cancers. CONCLUSION: PDX-1 is involved in the mechanisms of precancerous and cancerous lesions of the stomach and its overexpression is detected in the majority of the most malignant NETs and gastric cancers.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio , Tumores Neuroendócrinos , Neoplasias Gástricas , Transativadores , Humanos , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Feminino , Masculino , Pessoa de Meia-Idade , Transativadores/metabolismo , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Adulto , Idoso
3.
Dev Biol ; 477: 232-240, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34097879

RESUMO

In mammals, multiple cell-signaling pathways and transcription factors regulate development of the embryonic taste system and turnover of taste cells in the adult stage. Using single-cell RNA-Seq of mouse taste cells, we found that the homeobox-containing transcription factor Nkx2-2, a target of the Sonic Hedgehog pathway and a key regulator of the development and regeneration of multiple cell types in the body, is highly expressed in type III taste cells but not in type II or taste stem cells. Using in situ hybridization and immunostaining, we confirmed that Nkx2-2 is expressed specifically in type III taste cells in the endoderm-derived circumvallate and foliate taste papillae but not in the ectoderm-derived fungiform papillae. Lineage tracing revealed that Nkx2-2-expressing cells differentiate into type III, but not type II or type I cells in circumvallate and foliate papillae. Neonatal Nkx2-2-knockout mice did not express key type III taste cell marker genes, while the expression of type II and type I taste cell marker genes were unaffected in these mice. Our findings indicate that Nkx2-2-expressing cells are committed to the type III lineage and that Nkx2-2 may be critical for the development of type III taste cells in the posterior tongue, thus illustrating a key difference in the mechanism of type III cell lineage specification between ectoderm- and endoderm-derived taste fields.


Assuntos
Linhagem da Célula/fisiologia , Proteínas de Homeodomínio/fisiologia , Papilas Gustativas/embriologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/fisiologia , Contagem de Células , Linhagem da Célula/genética , Feminino , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/biossíntese , Masculino , Camundongos , RNA-Seq , Papilas Gustativas/citologia , Papilas Gustativas/metabolismo , Proteínas de Peixe-Zebra/biossíntese
4.
Development ; 146(15)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31371378

RESUMO

The spatiotemporal identity of neural progenitors and the regional control of neurogenesis are essential for the development of cerebral cortical architecture. Here, we report that mammalian DM domain factors (Dmrt) determine the identity of cerebral cortical progenitors. Among the Dmrt family genes expressed in the developing dorsal telencephalon, Dmrt3 and Dmrta2 show a medialhigh/laterallow expression gradient. Their simultaneous loss confers a ventral identity to dorsal progenitors, resulting in the ectopic expression of Gsx2 and massive production of GABAergic olfactory bulb interneurons in the dorsal telencephalon. Furthermore, double-mutant progenitors in the medial region exhibit upregulated Pax6 and more lateral characteristics. These ventral and lateral shifts in progenitor identity depend on Dmrt gene dosage. We also found that Dmrt factors bind to Gsx2 and Pax6 enhancers to suppress their expression. Our findings thus reveal that the graded expression of Dmrt factors provide positional information for progenitors by differentially repressing downstream genes in the developing cerebral cortex.


Assuntos
Córtex Cerebral/embriologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição PAX6/biossíntese , Fator de Transcrição PAX6/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética
5.
Nature ; 540(7633): 428-432, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27919074

RESUMO

The functionality of stem cells declines during ageing, and this decline contributes to ageing-associated impairments in tissue regeneration and function. Alterations in developmental pathways have been associated with declines in stem-cell function during ageing, but the nature of this process remains poorly understood. Hox genes are key regulators of stem cells and tissue patterning during embryogenesis with an unknown role in ageing. Here we show that the epigenetic stress response in muscle stem cells (also known as satellite cells) differs between aged and young mice. The alteration includes aberrant global and site-specific induction of active chromatin marks in activated satellite cells from aged mice, resulting in the specific induction of Hoxa9 but not other Hox genes. Hoxa9 in turn activates several developmental pathways and represents a decisive factor that separates satellite cell gene expression in aged mice from that in young mice. The activated pathways include most of the currently known inhibitors of satellite cell function in ageing muscle, including Wnt, TGFß, JAK/STAT and senescence signalling. Inhibition of aberrant chromatin activation or deletion of Hoxa9 improves satellite cell function and muscle regeneration in aged mice, whereas overexpression of Hoxa9 mimics ageing-associated defects in satellite cells from young mice, which can be rescued by the inhibition of Hoxa9-targeted developmental pathways. Together, these data delineate an altered epigenetic stress response in activated satellite cells from aged mice, which limits satellite cell function and muscle regeneration by Hoxa9-dependent activation of developmental pathways.


Assuntos
Senescência Celular , Epistasia Genética , Crescimento e Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Estresse Fisiológico/genética , Envelhecimento , Animais , Senescência Celular/genética , Cromatina/genética , Cromatina/metabolismo , Feminino , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Regeneração/genética
6.
Cereb Cortex ; 31(7): 3194-3212, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33675359

RESUMO

Thalamocortical neurons (TCNs) play a critical role in the maintenance of thalamocortical oscillations, dysregulation of which can result in certain types of seizures. Precise control over firing rates of TCNs is foundational to these oscillations, yet the transcriptional mechanisms that constrain these firing rates remain elusive. We hypothesized that Shox2 is a transcriptional regulator of ion channels important for TCN function and that loss of Shox2 alters firing frequency and activity, ultimately perturbing thalamocortical oscillations into an epilepsy-prone state. In this study, we used RNA sequencing and quantitative PCR of control and Shox2 knockout mice to determine Shox2-affected genes and revealed a network of ion channel genes important for neuronal firing properties. Protein regulation was confirmed by Western blotting, and electrophysiological recordings showed that Shox2 KO impacted the firing properties of a subpopulation of TCNs. Computational modeling showed that disruption of these conductances in a manner similar to Shox2's effects modulated frequency of oscillations and could convert sleep spindles to near spike and wave activity, which are a hallmark for absence epilepsy. Finally, Shox2 KO mice were more susceptible to pilocarpine-induced seizures. Overall, these results reveal Shox2 as a transcription factor important for TCN function in adult mouse thalamus.


Assuntos
Potenciais de Ação/fisiologia , Córtex Cerebral/metabolismo , Proteínas de Homeodomínio/biossíntese , Neurônios/metabolismo , Convulsões/metabolismo , Tálamo/metabolismo , Animais , Proteínas de Homeodomínio/genética , Canais Iônicos/biossíntese , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Rede Nervosa/metabolismo , Convulsões/genética , Convulsões/prevenção & controle , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
7.
Dev Biol ; 463(2): 110-123, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32422142

RESUMO

We show for the first time endoskeletal regeneration in the developing pectoral fin of zebrafish. The developing pectoral fin contains an aggregation plate of differentiated chondrocytes (endochondral disc; primordium for endoskeletal components, proximal radials). The endochondral disc can be regenerated after amputation in the middle of the disc. The regenerated disc sufficiently forms endoskeletal patterns. Early in the process of regenerating the endochondral disc, epithelium with apical ectodermal ridge (AER) marker expression rapidly covers the amputation plane, and mesenchymal cells start to actively proliferate. Taken together with re-expression of a blastema marker gene, msxb, and other developmental genes, it is likely that regeneration of the endochondral disc recaptures fin development as epimorphic limb regeneration does. The ability of endoskeletal regeneration declines during larval growth, and adult zebrafish eventually lose the ability to regenerate endoskeletal components such that amputated endoskeletons become enlarged. Endoskeletal regeneration in the zebrafish pectoral fin will serve as a new model system for successful appendage regeneration in mammals.


Assuntos
Nadadeiras de Animais/fisiologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Regeneração , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/metabolismo , Animais , Proteínas de Homeodomínio/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
Dev Biol ; 467(1-2): 39-50, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32891623

RESUMO

The Six1 transcription factor plays a major role in craniofacial development. Mutations in SIX1 and its co-factor, EYA1, are causative for about 50% of Branchio-otic/Branchio-oto-renal syndrome (BOR) patients, who are characterized by variable craniofacial, otic and renal malformations. We previously screened for other proteins that might interact with Six1 to identify additional genes that may play a role in BOR, and herein characterize the developmental role of one of them, Microspherule protein 1 (Mcrs1). We found that in cultured cells, Mcrs1 bound to Six1 and in both cultured cells and embryonic ectoderm reduced Six1-Eya1 transcriptional activation. Knock-down of Mcrs1 in embryos caused an expansion of the domains of neural plate genes and two genes expressed in both the neural plate and neural crest (zic1, zic2). In contrast, two other genes expressed in pre-migratory neural crest (foxd3, sox9) were primarily reduced. Cranial placode genes showed a mixture of expanded and diminished expression domains. At larval stages, loss of Mcrs1 resulted in a significant reduction of otic vesicle gene expression concomitant with a smaller otic vesicle volume. Experimentally increasing Mcrs1 above endogenous levels favored the expansion of neural border and neural crest gene domains over cranial placode genes; it also reduced otic vesicle gene expression but not otic vesicle volume. Co-expression of Mcrs1 and Six1 as well as double knock-down and rescue experiments establish a functional interaction between Mcrs1 and Six1 in the embryo, and demonstrate that this interaction has an important role in the development of craniofacial tissues including the otic vesicle.


Assuntos
Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Ligação a RNA/biossíntese , Crânio/embriologia , Proteínas de Xenopus/biossíntese , Animais , Ectoderma/embriologia , Crista Neural/embriologia , Xenopus laevis
9.
J Biol Chem ; 295(34): 12188-12202, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32467227

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive cancer subtype for which effective therapies are unavailable. TNBC has a high frequency of tumor protein p53 (Tp53/p53)- and phosphatase and tensin homolog (PTEN) deficiencies, and combined p53- and PTEN-deficiency is associated with poor prognosis and poor response to anticancer therapies. In this study, we discovered that combined p53- and PTEN-deficiency in TNBC activates expression of the transcription factor mesenchyme homeobox 1 (MEOX1). We found that MEOX1 is expressed only in TNBC cells with frequent deficiencies in p53 and PTEN, and that its expression is undetectable in luminal A, luminal B, and HER2+ subtypes, as well as in normal breast cells with wild-type (WT) p53 and PTEN. Notably, siRNA knockdown of both p53 and PTEN activated MEOX1 expression in breast cancer cells, whereas individual knockdowns of either p53 or PTEN had only minimal effects on MEOX1 expression. MEOX1 knockdown abolished cell proliferation of p53- and PTEN-deficient TNBC in vitro and inhibited tumor growth in vivo, but had no effect on the proliferation of luminal and HER2+ cancer cells and normal breast cells. RNA-Seq and immunoblotting analyses showed that MEOX1 knockdown decreased expression of tyrosine kinase 2 (TYK2), signal transducer and activator of transcription 5B (STAT5B), and STAT6 in p53- and PTEN-deficient TNBC cells. These results reveal the effects of combined p53- and PTEN-deficiency on MEOX1 expression and TNBC cell proliferation, suggesting that MEOX1 may serve as a potential therapeutic target for managing p53- and PTEN-deficient TNBC.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , PTEN Fosfo-Hidrolase/deficiência , Fatores de Transcrição/biossíntese , Proteína Supressora de Tumor p53/deficiência , Animais , Feminino , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , PTEN Fosfo-Hidrolase/metabolismo , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT6/biossíntese , Fator de Transcrição STAT6/genética , TYK2 Quinase/biossíntese , TYK2 Quinase/genética , Fatores de Transcrição/genética , Neoplasias de Mama Triplo Negativas , Proteína Supressora de Tumor p53/metabolismo
10.
J Biol Chem ; 295(37): 12975-12992, 2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32690606

RESUMO

Pancreas/duodenum homeobox protein 1 (PDX1) is an important transcription factor that regulates islet ß-cell proliferation, differentiation, and function. Reduced expression of PDX1 is thought to contribute to ß-cell loss and dysfunction in diabetes. Thus, promoting PDX1 expression can be an effective strategy to preserve ß-cell mass and function. Previously, we established a PDX1 promoter-dependent luciferase system to screen agents that can promote PDX1 expression. Natural compound tectorigenin (TG) was identified as a promising candidate that could enhance the activity of the promoter for the PDX1 gene. In this study, we first demonstrated that TG could promote the expression of PDX1 in ß-cells via activating extracellular signal-related kinase (ERK), as indicated by increased phosphorylation of ERK; this effect was observed under either normal or glucotoxic/lipotoxic conditions. We then found that TG could suppress induced apoptosis and improved the viability of ß-cells under glucotoxicity and lipotoxicity by activation of ERK and reduction of reactive oxygen species and endoplasmic reticulum (ER) stress. These effects held true in vivo as well: prophylactic or therapeutic use of TG could obviously inhibit ER stress and decrease islet ß-cell apoptosis in the pancreas of mice given a high-fat/high-sucrose diet (HFHSD), thus dramatically maintaining or restoring ß-cell mass and islet size, respectively. Accordingly, both prophylactic and therapeutic use of TG improved HFHSD-impaired glucose metabolism in mice, as evidenced by ameliorating hyperglycemia and glucose intolerance. Taken together, TG, as an agent promoting PDX1 expression exhibits strong protective effects on islet ß-cells both in vitro and in vivo.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/biossíntese , Células Secretoras de Insulina/metabolismo , Isoflavonas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Regiões Promotoras Genéticas , Transativadores/biossíntese , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Glucose/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Ratos
11.
Hum Mol Genet ; 28(3): 476-486, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30312408

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is a common, dominantly inherited disease caused by the epigenetic de-repression of the DUX4 gene, a transcription factor normally repressed in skeletal muscle. As targeted therapies are now possible in FSHD, a better understanding of the relationship between DUX4 activity, muscle pathology and muscle magnetic resonance imaging (MRI) changes is crucial both to understand disease mechanisms and for the design of future clinical trials. Here, we performed MRIs of the lower extremities in 36 individuals with FSHD, followed by needle muscle biopsies in safely accessible muscles. We examined the correlation between MRI characteristics, muscle pathology and expression of DUX4 target genes. Results show that the presence of elevated MRI short tau inversion recovery signal has substantial predictive value in identifying muscles with active disease as determined by histopathology and DUX4 target gene expression. In addition, DUX4 target gene expression was detected only in FSHD-affected muscles and not in control muscles. These results support the use of MRI to identify FSHD muscles most likely to have active disease and higher levels of DUX4 target gene expression and might be useful in early phase therapeutic trials to demonstrate target engagement in therapies aiming to suppress DUX4 expression.


Assuntos
Proteínas de Homeodomínio/genética , Músculo Esquelético/patologia , Distrofia Muscular Facioescapuloumeral/diagnóstico por imagem , Adulto , Idoso , Biópsia , Feminino , Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Fatores de Transcrição/genética
12.
Br J Cancer ; 124(4): 805-816, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33214683

RESUMO

BACKGROUND: Understanding the pathways that drive adrenocortical carcinoma (ACC) is essential to the development of more effective therapies. This study investigates the role of the transcription factor HOXB9 and other HOX factors in ACC and its treatment. METHODS: We used transgenic mouse models to determine the role of Hoxb9 in adrenal tumour development. Patient transcriptomic data was analysed for the expression of HOX genes and their association with disease. Drug response studies on various adrenocortical models were done to establish novel therapeutic options. RESULTS: Our human ACC dataset analyses showed high expression of HOXB9, and other HOX factors, are associated with poorer prognosis. Transgenic overexpression of Hoxb9 in the adrenal cortex of mice with activated Ctnnb1 led to larger adrenal tumours. This phenotype was preferentially observed in male mice and was characterised by more proliferating cells and an increase in the expression of cell cycle genes, including Ccne1. Adrenal tumour cells were found to be dependent on HOX function for survival and were sensitive to a specific peptide inhibitor. CONCLUSIONS: These studies show Hoxb9 can promote adrenal tumour progression in a sex-dependent manner and have identified HOX factors as potential drug targets, leading to novel therapeutic approaches in ACC.


Assuntos
Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Neoplasias do Córtex Suprarrenal/genética , Carcinoma Adrenocortical/tratamento farmacológico , Carcinoma Adrenocortical/genética , Proteínas de Homeodomínio/genética , Peptídeos/farmacologia , Neoplasias do Córtex Suprarrenal/patologia , Carcinoma Adrenocortical/patologia , Animais , Proliferação de Células/genética , Feminino , Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Terapia de Alvo Molecular , Peptídeos/genética
13.
J Cell Sci ; 132(2)2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30630896

RESUMO

Chromatin assembly factor 1 (CAF1), a histone chaperone that mediates the deposition of histone H3/H4 onto newly synthesized DNA, is involved in Notch signaling activation during Drosophila wing imaginal disc development. Here, we report another side of CAF1, wherein the subunits CAF1-p105 and CAF1-p180 (also known as CAF1-105 and CAF1-180, respectively) inhibit expression of Notch target genes and show this is required for proliferation of Drosophila ovarian follicle cells. Loss-of-function of either CAF1-p105 or CAF1-p180 caused premature activation of Notch signaling reporters and early expression of the Notch target Hindsight (Hnt, also known as Pebbled), leading to Cut downregulation and inhibition of follicle cell mitosis. Our studies further show Notch is functionally responsible for these phenotypes observed in both the CAF1-p105- and CAF1-p180-deficient follicle cells. Moreover, we reveal that CAF1-p105- and CAF1-p180-dependent Cut expression is essential for inhibiting Hnt expression in follicle cells during their mitotic stage. These findings together indicate a novel negative-feedback regulatory loop between Cut and Hnt underlying CAF1-p105 and CAF-p180 regulation, which is crucial for follicle cell differentiation. In conclusion, our studies suggest CAF1 plays a dual role to sustain cell proliferation by positively or negatively regulating Drosophila Notch signaling in a tissue-context-dependent manner.


Assuntos
Proliferação de Células , Proteínas de Drosophila/metabolismo , Folículo Ovariano/metabolismo , Receptores Notch/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Transdução de Sinais , Animais , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Discos Imaginais/citologia , Discos Imaginais/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Folículo Ovariano/citologia , Receptores Notch/genética , Proteína 4 de Ligação ao Retinoblastoma/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
14.
Development ; 145(13)2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29945867

RESUMO

Neuroendocrine cells in the pineal gland release melatonin during the night and, in teleosts, are directly photoreceptive. During development of the pineal complex, a small number of cells migrate leftward away from the pineal anlage to form the parapineal cell cluster, a process that is crucial for asymmetrical development of the bilateral habenular nuclei. Here, we show that, throughout zebrafish embryonic development, the brain-specific homeobox (bsx) gene is expressed in all cell types of the pineal complex. We identified Bmp and Noto/Flh as major regulators of bsx expression in the pineal complex. Upon loss of Bsx through the generation of a targeted mutation, embryos fail to form a parapineal organ and develop right-isomerized habenulae. Crucial enzymes in the melatonin biosynthesis pathway are not expressed, suggesting the absence of melatonin from the pineal gland in bsx mutants. Several genes involved in rod-like or cone-like phototransduction are also abnormally expressed, indicating that Bsx has a pivotal role in the differentiation of multiple cell types in the zebrafish pineal complex.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/biossíntese , Glândula Pineal/embriologia , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/embriologia , Animais , Proteínas de Homeodomínio/genética , Melatonina/biossíntese , Melatonina/genética , Glândula Pineal/citologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
Development ; 145(13)2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29853618

RESUMO

Although the specific form of an organ is frequently important for its function, the mechanisms underlying organ shape are largely unknown. In Drosophila, the wings and halteres, homologous appendages of the second and third thoracic segments, respectively, bear different forms: wings are flat, whereas halteres are globular, and yet both characteristic shapes are essential for a normal flight. The Hox gene Ultrabithorax (Ubx) governs the difference between wing and haltere development, but how Ubx function in the appendages prevents or allows flat or globular shapes is unknown. Here, we show that Ubx downregulates Matrix metalloproteinase 1 (Mmp1) expression in the haltere pouch at early pupal stage, which in turn prevents the rapid clearance of Collagen IV compared with the wing disc. This difference is instrumental in determining cell shape changes, expansion of the disc and apposition of dorsal and ventral layers, all of these phenotypic traits being characteristic of wing pouch development. Our results suggest that Ubx regulates organ shape by controlling Mmp1 expression, and the extent and timing of extracellular matrix degradation.


Assuntos
Proteínas de Drosophila/biossíntese , Matriz Extracelular/metabolismo , Proteínas de Homeodomínio/biossíntese , Discos Imaginais/embriologia , Metaloproteinase 1 da Matriz/metabolismo , Fatores de Transcrição/biossíntese , Asas de Animais/embriologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Matriz Extracelular/genética , Proteínas de Homeodomínio/genética , Metaloproteinase 1 da Matriz/genética , Fatores de Transcrição/genética
16.
Development ; 145(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29229773

RESUMO

The evolution of a hinged moveable jaw with variable morphology is considered a major factor behind the successful expansion of the vertebrates. DLX homeobox transcription factors are crucial for establishing the positional code that patterns the mandible, maxilla and intervening hinge domain, but how the genes encoding these proteins are regulated remains unclear. Herein, we demonstrate that the concerted action of the AP-2α and AP-2ß transcription factors within the mouse neural crest is essential for jaw patterning. In the absence of these two proteins, the hinge domain is lost and there are alterations in the size and patterning of the jaws correlating with dysregulation of homeobox gene expression, with reduced levels of Emx, Msx and Dlx paralogs accompanied by an expansion of Six1 expression. Moreover, detailed analysis of morphological features and gene expression changes indicate significant overlap with various compound Dlx gene mutants. Together, these findings reveal that the AP-2 genes have a major function in mammalian neural crest development, influencing patterning of the craniofacial skeleton via the DLX code, an effect that has implications for vertebrate facial evolution, as well as for human craniofacial disorders.


Assuntos
Padronização Corporal/fisiologia , Região Branquial/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/biossíntese , Crista Neural/metabolismo , Fator de Transcrição AP-2/metabolismo , Animais , Região Branquial/citologia , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Crista Neural/citologia , Fator de Transcrição AP-2/genética
17.
Development ; 145(14)2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-29967281

RESUMO

Dopamine receptor DRD1-expressing medium spiny neurons (D1 MSNs) and dopamine receptor DRD2-expressing medium spiny neurons (D2 MSNs) are the principal projection neurons in the striatum, which is divided into dorsal striatum (caudate nucleus and putamen) and ventral striatum (nucleus accumbens and olfactory tubercle). Progenitors of these neurons arise in the lateral ganglionic eminence (LGE). Using conditional deletion, we show that mice lacking the transcription factor genes Sp8 and Sp9 lose virtually all D2 MSNs as a result of reduced neurogenesis in the LGE, whereas D1 MSNs are largely unaffected. SP8 and SP9 together drive expression of the transcription factor Six3 in a spatially restricted domain of the LGE subventricular zone. Conditional deletion of Six3 also prevents the formation of most D2 MSNs, phenocopying the Sp8/9 mutants. Finally, ChIP-Seq reveals that SP9 directly binds to the promoter and a putative enhancer of Six3 Thus, this study defines components of a transcription pathway in a regionally restricted LGE progenitor domain that selectively drives the generation of D2 MSNs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/citologia , Neurônios/citologia , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Fatores de Transcrição/genética , Proteína Homeobox SIX3
18.
Development ; 145(11)2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29752384

RESUMO

During zebrafish fin regeneration, blastema cells lining the epidermis differentiate into osteoblasts and joint cells to reconstruct the segmented bony rays. We show that osteoblasts and joint cells originate from a common cell lineage, but are committed to different cell fates. Pre-osteoblasts expressing runx2a/b commit to the osteoblast lineage upon expressing sp7, whereas the strong upregulation of hoxa13a correlates with a commitment to a joint cell type. In the distal regenerate, hoxa13a, evx1 and pthlha are sequentially upregulated at regular intervals to define the newly identified presumptive joint cells. Presumptive joint cells mature into joint-forming cells, a distinct cell cluster that maintains the expression of these factors. Analysis of evx1 null mutants reveals that evx1 is acting upstream of pthlha and downstream of or in parallel with hoxa13a Calcineurin activity, potentially through the inhibition of retinoic acid signaling, regulates evx1, pthlha and hoxa13a expression during joint formation. Furthermore, retinoic acid treatment induces osteoblast differentiation in mature joint cells, leading to ectopic bone deposition in joint regions. Overall, our data reveal a novel regulatory pathway essential for joint formation in the regenerating fin.


Assuntos
Nadadeiras de Animais/crescimento & desenvolvimento , Calcineurina/metabolismo , Articulações/crescimento & desenvolvimento , Regeneração/fisiologia , Tretinoína/farmacologia , Peixe-Zebra/fisiologia , Animais , Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Osteoblastos/citologia , Proteína Relacionada ao Hormônio Paratireóideo/biossíntese , Proteína Relacionada ao Hormônio Paratireóideo/genética , Fator de Transcrição Sp7/biossíntese , Fator de Transcrição Sp7/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
19.
Development ; 145(17)2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30177510

RESUMO

During early embryonic development of the spinal cord, graded sonic hedgehog signaling establishes distinct ventral progenitor domains by regulating the spatiotemporal expression of fate-specifying transcription factors. However, regulation of their protein stability remains incompletely understood. Here, we show that RNF220, an E3 ubiquitin ligase, plays crucial roles in the generation of the ventral progenitor domains, which produce ventral interneurons and motor neurons, by targeting key transcription factors including Dbx1/2 and Nkx2.2 for degradation. Surprisingly, RNF220 interacts with, and is co-expressed with, a zinc-finger protein ZC4H2, and they cooperate to degrade Dbx1/2 and Nkx2.2. RNF220-null mice show widespread alterations of ventral progenitor domains, including the loss of the p2 domain that produces V2 interneurons. Knockdown of RNF220 and ZC4H2 in the chick spinal cord downregulates expression of the V2 interneuronal marker Chx10. Co-expression of RNF220 and ZC4H2 further promotes the ability of Nkx6.1 to induce ectopic Chx10+ V2 interneurons. Our results uncover a novel regulatory pathway in establishing distinct progenitor domains through modulating the protein stability of transcription factors. Our results provide insights into the molecular mechanism by which ZC4H2 mutations lead to human syndromes characterized by delayed motor development.


Assuntos
Interneurônios/citologia , Neurônios Motores/citologia , Células-Tronco Neurais/citologia , Proteínas Nucleares/metabolismo , Medula Espinal/embriologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Embrião de Galinha , Células HEK293 , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/genética , Peixe-Zebra , Proteínas de Peixe-Zebra
20.
Exp Eye Res ; 209: 108629, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34029595

RESUMO

The shortage of human donor corneas has raised important concerns about engineering of corneal endothelial cells (CECs) for clinical use. However, due to the limited proliferative capacity of human CECs, driving them into proliferation and regeneration may be difficult. Unlike human CECs, rabbit CECs have a marked proliferative capacity. To clarify the potential reason for this difference, we analysed the proteomes of four human corneal endothelium samples and four rabbit corneal endothelium samples with quantitative label-free proteomics and downstream analysis. We discovered that vitamin and selenocompound metabolism and some signaling pathways such as NF-kappa B signaling pathway differed between the samples. Moreover, TGFß, PITX2 and keratocan were distinctively expressed in rabbit samples, which might be associated with active proliferation in rabbit CECs. This study illustrates the proteomic differences between human and rabbit CECs and might promote CEC engineering strategies.


Assuntos
Doenças da Córnea/metabolismo , Transplante de Córnea/métodos , Endotélio Corneano/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteoma/metabolismo , Proteômica/métodos , Fatores de Transcrição/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Idoso , Animais , Diferenciação Celular , Células Cultivadas , Doenças da Córnea/patologia , Doenças da Córnea/cirurgia , Modelos Animais de Doenças , Endotélio Corneano/citologia , Endotélio Corneano/transplante , Humanos , Masculino , Pessoa de Meia-Idade , Coelhos , Preservação de Tecido/métodos , Proteína Homeobox PITX2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA