Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 810
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(16): e2316244121, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38588419

RESUMO

Despite the conservation of genetic machinery involved in eye development, there is a strong diversity in the placement of eyes on the head of animals. Morphogen gradients of signaling molecules are vital to patterning cues. During Drosophila eye development, Wingless (Wg), a ligand of Wnt/Wg signaling, is expressed anterolaterally to form a morphogen gradient to determine the eye- versus head-specific cell fate. The underlying mechanisms that regulate this process are yet to be fully understood. We characterized defective proventriculus (dve) (Drosophila ortholog of human SATB1), a K50 homeodomain transcription factor, as a dorsal eye gene, which regulates Wg signaling to determine eye versus head fate. Across Drosophila species, Dve is expressed in the dorsal head vertex region where it regulates wg transcription. Second, Dve suppresses eye fate by down-regulating retinal determination genes. Third, the dve-expressing dorsal head vertex region is important for Wg-mediated inhibition of retinal cell fate, as eliminating the Dve-expressing cells or preventing Wg transport from these dve-expressing cells leads to a dramatic expansion of the eye field. Together, these findings suggest that Dve regulates Wg expression in the dorsal head vertex, which is critical for determining eye versus head fate. Gain-of-function of SATB1 exhibits an eye fate suppression phenotype similar to Dve. Our data demonstrate a conserved role for Dve/SATB1 in the positioning of eyes on the head and the interocular distance by regulating Wg. This study provides evidence that dysregulation of the Wg morphogen gradient results in developmental defects such as hypertelorism in humans where disproportionate interocular distance and facial anomalies are reported.


Assuntos
Proteínas de Drosophila , Proteínas de Ligação à Região de Interação com a Matriz , Animais , Humanos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Drosophila/genética , Retina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Drosophila melanogaster/metabolismo , Padronização Corporal/genética
2.
Nat Commun ; 14(1): 6174, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37798281

RESUMO

The control of Wnt receptor abundance is critical for animal development and to prevent tumorigenesis, but the mechanisms that mediate receptor stabilization remain uncertain. We demonstrate that stabilization of the essential Wingless/Wnt receptor Arrow/LRP6 by the evolutionarily conserved Usp46-Uaf1-Wdr20 deubiquitylase complex controls signaling strength in Drosophila. By reducing Arrow ubiquitylation and turnover, the Usp46 complex increases cell surface levels of Arrow and enhances the sensitivity of target cells to stimulation by the Wingless morphogen, thereby increasing the amplitude and spatial range of signaling responses. Usp46 inactivation in Wingless-responding cells destabilizes Arrow, reduces cytoplasmic accumulation of the transcriptional coactivator Armadillo/ß-catenin, and attenuates or abolishes Wingless target gene activation, which prevents the concentration-dependent regulation of signaling strength. Consequently, Wingless-dependent developmental patterning and tissue homeostasis are disrupted. These results reveal an evolutionarily conserved mechanism that mediates Wnt/Wingless receptor stabilization and underlies the precise activation of signaling throughout the spatial range of the morphogen gradient.


Assuntos
Proteínas de Drosophila , Via de Sinalização Wnt , Animais , Proteínas de Drosophila/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Drosophila/genética , Fatores de Transcrição/metabolismo
3.
Int J Mol Sci ; 24(9)2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37176048

RESUMO

Endometrial cancer remains a common cancer affecting the female reproductive system. There is still a need for more efficient ways of determining the degree of malignancy and optimizing treatment. WNT and mTOR are components of signaling pathways within tumor cells, and dysfunction of either protein is associated with the pathogenesis of neoplasms. Therefore, the aim of our study was to assess the impact of subcellular WNT-1 and mTOR levels on the clinical course of endometrial cancer. WNT-1 and mTOR levels in the plasma membrane, nucleus, and cytoplasm were evaluated using immunohistochemical staining in a group of 64 patients with endometrial cancer of grades 1-3 and FIGO stages I-IV. We discovered that the levels of WNT-1 and mTOR expression in the cellular compartments were associated with tumor grade and staging. Membranous WNT-1 was negatively associated, whereas cytoplasmic WNT-1 and nuclear mTOR were positively associated with higher grading of endometrial cancer. Furthermore, nuclear mTOR was positively associated with FIGO stages IB-IV. To conclude, we found that the assessment of WNT-1 in the cell membrane may be useful for exclusion of grade 3 neoplasms, whereas cytoplasmic WNT-1 and nuclear mTOR may be used as indicators for confirmation of grade 3 neoplasms.


Assuntos
Neoplasias do Endométrio , Feminino , Humanos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Estadiamento de Neoplasias , Serina-Treonina Quinases TOR/genética , Proteína Wnt1/metabolismo
4.
Biol Open ; 12(5)2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37199309

RESUMO

Cell recruitment is a process by which a differentiated cell induces neighboring cells to adopt its same cell fate. In Drosophila, cells expressing the protein encoded by the wing selector gene, vestigial (vg), drive a feed-forward recruitment signal that expands the Vg pattern as a wave front. However, previous studies on Vg pattern formation do not reveal these dynamics. Here, we use live imaging to show that multiple cells at the periphery of the wing disc simultaneously activate a fluorescent reporter of the recruitment signal, suggesting that cells may be recruited without the need for their contact neighbors be recruited in advance. In support of this observation, when Vg expression is inhibited either at the dorsal-ventral boundary or away from it, the activation of the recruitment signal still occurs at a distance, suggesting that Vg expression is not absolutely required to send or propagate the recruitment signal. However, the strength and extent of the recruitment signal is clearly compromised. We conclude that a feed-forward, contact-dependent cell recruitment process is not essential for Vg patterning, but it is necessary for robustness. Overall, our findings reveal a previously unidentified role of cell recruitment as a robustness-conferring cell differentiation mechanism.


Assuntos
Proteínas de Drosophila , Drosophila , Proteínas Nucleares , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
5.
J Cell Sci ; 136(7)2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36897575

RESUMO

Morphogens provide quantitative and robust signaling systems to achieve stereotypic patterning and morphogenesis. Heparan sulfate (HS) proteoglycans (HSPGs) are key components of such regulatory feedback networks. In Drosophila, HSPGs serve as co-receptors for a number of morphogens, including Hedgehog (Hh), Wingless (Wg), Decapentaplegic (Dpp) and Unpaired (Upd, or Upd1). Recently, Windpipe (Wdp), a chondroitin sulfate (CS) proteoglycan (CSPG), was found to negatively regulate Upd and Hh signaling. However, the roles of Wdp, and CSPGs in general, in morphogen signaling networks are poorly understood. We found that Wdp is a major CSPG with 4-O-sulfated CS in Drosophila. Overexpression of wdp modulates Dpp and Wg signaling, showing that it is a general regulator of HS-dependent pathways. Although wdp mutant phenotypes are mild in the presence of morphogen signaling buffering systems, this mutant in the absence of Sulf1 or Dally, molecular hubs of the feedback networks, produces high levels of synthetic lethality and various severe morphological phenotypes. Our study indicates a close functional relationship between HS and CS, and identifies the CSPG Wdp as a novel component in morphogen feedback pathways.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Proteoglicanas de Sulfatos de Condroitina/genética , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Sulfatases/genética , Sulfatases/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
6.
Sci Rep ; 12(1): 14934, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056132

RESUMO

Wnt1 is the first mammalian Wnt gene, which is discovered as proto-oncogene and in human the gene is located on the chromosome 12q13. Mutations in Wnt1 are reported to be associated with various cancers and other human diseases. The structural and functional consequences of most of the non-synonymous SNPs (nsSNPs), present in the human Wnt1 gene, are not known. In the present work, extensive bioinformatics analyses are used to screen 292 nsSNPs of Wnt1 for predicting pathogenic and harmless polymorphisms. We have identified 10 highly deleterious nsSNPs among which 7 are located within the highly conserved areas. These 10 nsSNPs are also predicted to affect the post-translational modifications of Wnt1. Further, structure based stability analyses of these 10 highly deleterious nsSNPs revealed 8 variants as highly destabilizing. These 8 highly destabilizing variants were shown to have high BC score and high RMSIP score from normal mode analyses. Based on the deformation energies, obtained from the normal mode analyses, variants like G169A, G169S, G331R and G331S were found to be unstable. Molecular Dynamics (MD) simulations revealed structural stability and fluctuation of WT Wnt1 and its prioritized variants. RMSD remained fluctuating mostly between 4 and 5 Å and occasionally between 3.5 and 5.5 Å ranges. RMSF in the CTD region (residues 330-360) of the binding pocket were lower compared to that of WT. Studying the impacts of nsSNPs on the binding interface of Wnt1 and seven Frizzled receptors have predicted substitutions which can stabilize or destabilize the binding interface. We have found that Wnt1 and FZD8-CRD is the best docked complex in our study. MD simulation based analyses of wild type Wnt1-FZD8-CRD complex and the 8 prioritized variants revealed that RMSF was higher in the unstructured regions and RMSD remained fluctuating in the region of 5 Å ± 1 Å. We have also observed differential Wnt1 gene expression pattern in normal, tumor and metastatic conditions across different tissues. Wnt1 gene expression was significantly higher in metastatic tissues of lungs, colon and skin; and was significantly lower in metastatic tissues of breast, esophagus and kidney. We have also found that Wnt1 deregulation is associated with survival outcome in patients with gastric and breast cancer. Furthermore, these computationally screened highly deleterious nsSNPs of Wnt1 can be analyzed in population based genetic studies and may help understand the Wnt1 associated diseases.


Assuntos
Receptores Frizzled , Polimorfismo de Nucleotídeo Único , Proteína Wnt1/genética , Carcinogênese , Biologia Computacional , Receptores Frizzled/genética , Humanos , Simulação de Dinâmica Molecular , Proteína Wnt1/química , Proteína Wnt1/metabolismo
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(8): 692-698, 2022 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-35851082

RESUMO

Objective To explore the effect and mechanism of paeoniflorin (PAE) on apoptosis of fibroblast synovial cells derived from rheumatoid arthritis tissues. Methods Rheumatoid arthritis fibroblast-like synovial cells (RA-FLSs) were cultured under different concentrations of PAE (20, 40, 80 µmol/L). Survival rate of cells was determined at different incubation time-points (24, 48, 72 hours) by MTT assay. Flow cytometry was performed to determine apoptosis rate of cells. Cells were transfected with 3 small interfering RNAs(siRNAs) fragments of long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1(lncRNA MALAT1) for 48 hours. Real-time quantitative PCR was performed to determine if MALAT1 knockdown was successful. Cells were assigned into control group, PAE group, si-NC group, si-MALAT1 group and PAE combined with si-MALAT1 group. Cell apoptosis rate was determined by flow cytometry after knockdown of MALAT1 expression. mRNA expression levels of Wnt1, ß-catenin, caspase-3, caspase-9, B-cell lymphoma 2(Bcl2) and Bcl2-associated X protein (BAX) were determined using real-time quantitative PCR. Western blot analysis was performed to determine protein expression levels of Wnt1, ß-catenin, caspase-3, caspase-9, Bcl2 and BAX in each group. Results The findings showed that incubation of RA-FLSs with PAE at the tested concentration decreased their viability in a time-dependent manner. Expression level of lncRNA MALAT1 was lower in RA-FLSs group compared with the level in NC-FLSs group. Analysis showed that apoptosis rate was higher in cells treated with PAE. si-MALAT1 group had the lowest apoptosis rate, whereas the PAE combined with si-MALAT1 group showed a significantly higher apoptosis rate compared with si-MALAT1 group. mRNA levels of Bcl2, Wnt1 and ß-catenin were lower in PAE group compared with the level in control group, whereas mRNA expression levels of caspase-3, caspase-9 and BAX were higher in PAE group compared with the levels in control group. Expression level of Bcl2 was significantly higher in si-MALAT1 group compared with the level in the control group. Expression level of BAX, caspase-3 and caspase-9 were higher in PAE combined with si-MALAT1 group compared with the level in the control group. Conclusion PAE promotes apoptosis and inhibits the Wnt1/ß-catenin pathway in RA-FLSs by upregulating expression of lncRNA MALAT1.


Assuntos
Artrite Reumatoide , RNA Longo não Codificante , Sinoviócitos , Apoptose/genética , Artrite Reumatoide/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Proliferação de Células/genética , Células Cultivadas , Fibroblastos/metabolismo , Glucosídeos , Humanos , Monoterpenos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Sinoviócitos/metabolismo , Via de Sinalização Wnt , Proteína Wnt1/metabolismo , Proteína X Associada a bcl-2/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-35675541

RESUMO

OBJECTIVE: To explore the role of WNT family member 1 (WNT1) in the development of dysplasia of the hip (DDH) and the molecular mechanism involved in this process. Methods: Si-WNT1, pcDNA3.1-WNT1 or corresponding negative controls were transfected into human osteoblast hFOB1.19 and human chondrocyte C28/I2, respectively. The proliferation of cells was measured by EdU assay. The relative expressions of human noggin gene (NOG), growth differentiating factor 5 (GDF5), WNT1, and WNT1-inducible-signaling pathway protein 2 (WISP2) were determined by immunofluorescence analysis. The protein expressions of RNA-binding protein of multiple splice forms 2 (RBPMS2), NOG, bone morphogenetic protein 2 (BMP2), BMP4, WNT1 and WISP2 were determined by western blot. Animal experiment was also performed and the morphological development of hip joint was observed. Results: Overexpression of WNT1 promoted osteoblast proliferation and inhibited chondrocyte proliferation, while knockdown of WNT1 inhibited osteoblast proliferation. In chondrocytes, knockdown of WNT1 upregulated NOG expression, while overexpression of WNT1 downregulated its expression. In osteoblasts and chondrocytes, overexpression of WNT1 increased BMP2, BMP4, WNT1, and WISP2 expression. RBPMS2 and NOG were slightly expressed in each group. Conclusion: Overexpression of WNT1 promoted osteoblast proliferation, inhibited chondrocyte proliferation, and increased the expressions of BMP2, BMP4, WNT1, and WISP2. Therefore, WNT1 may be a new therapeutic target for DDH.


Assuntos
Luxação Congênita de Quadril , Osteoblastos , Proteína Wnt1 , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Diferenciação Celular , Proliferação de Células , Fator V/metabolismo , Fator 5 de Diferenciação de Crescimento/metabolismo , Luxação Congênita de Quadril/metabolismo , Humanos , Osteoblastos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Proteína Wnt1/metabolismo
9.
Bioengineered ; 13(5): 12409-12420, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35603707

RESUMO

Intrauterine infection induces inflammation-mediated microglial activation and brain injury. This study aimed to explore the regulatory mechanism of Wnt family member 1 (Wnt1) in intrauterine infection-mediated microglial polarization. The cell counting kit-8 (CCK-8) assay was used to determine the viability of microglia, and cytokine expression levels were determined using enzyme linked immunosorbent assay (ELISA) kits and real-time quantitative PCR (RT-qPCR). The number of CD206+ and CD16/32+ cells was determined by flow cytometry. Wnt1 expression was analyzed using western blotting and immunofluorescence. Moreover, an in vivo assay was performed to verify the role of WNT1 in inflammation-sensitized brain injury in newborn mice. Lipopolysaccharide (LPS) exposure resulted in a decrease in microglial cell viability while increasing the expression levels of inflammatory cytokines (TNF-α, IL-6, and IL-1ß), simultaneously promoting M1-type microglial conversion. However, these effects were rescued by overexpression of Wnt1, which was expressed less in microglia exposed to LPS in vitro and in vivo. Here, we found that Wnt1 activated the LKB1-AMPK pathway, and the inhibition of LKB1 attenuated the rescue effects of Wnt1. In addition, LPS exposure reduced the autophagy of microglia, and Wnt1 overexpression enhanced the autophagy, but this effect was reversed by treatment with an LKB1 inhibitor. Wnt1 activated LKB1 to suppress inflammation-mediated activation of microglia, promote M2-type microglia conversion via the AMPK pathway, and alleviate inflammation-sensitized neonatal brain injuries. This provides a potential avenue for the treatment of neonatal brain injuries.


Assuntos
Lesões Encefálicas , Microglia , Proteína Wnt1/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Citocinas/metabolismo , Família , Inflamação/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/metabolismo , Transdução de Sinais
10.
Med Oncol ; 39(4): 42, 2022 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-35092507

RESUMO

Paranasal sinus tumors are a rare type of cancer. Most of these tumors are of epithelial origin and 80% of them are maxillary sinus squamous cell carcinoma. The WNT signaling pathway is an essential embryonic regulatory pathway known to play an important role in many cancers, including head and neck cancers. However, the effect of this pathway in maxillary sinus tumors has not been studied before. The aim of the study was to determine the changes in the regulatory genes of the WNT signaling pathway in maxillary sinus tumors. For this purpose, total RNA was isolated from the pathological preparations of 85 patients who had previously been operated on for squamous cell maxillary sinus tumor, and gene expression changes were evaluated by real-time RT-qPCR. The interactions among proteins encoded by genes, whose expression levels were found to be decreased and increased, were determined by protein-protein interaction (PPI) network analysis using string database, and signaling pathways that they are involved in were examined by Reactome database. A significant decrease in the expression of 28 genes compared to the control (fold change < 2.00 and p-value < 0.05) and a significant increase in the expression of 23 genes (fold change < 2.00 and p-value < 0.05) were detected. According to in silico analysis results, Signal Transduction (REACTOME:R-HSA-162582) and Signaling by WNT (REACTOME:R-HSA-195721) pathways were determined as most regulated pathways and FZD4-LRP5 and BCL9-CTNNB1 were determined as the strongest interactions. The current study contributes to illuminating the genetic regulation of maxillary sinus carcinoma in which genetic knowledge is limited. Our findings take attention to the dysregulations of the WNT signaling pathway that may support maxillary sinus carcinogenesis. The results will pave the way for further studies that investigate the therapy target potential of the WNT signaling pathway in this rare cancer.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias dos Seios Paranasais/patologia , Via de Sinalização Wnt/fisiologia , Proteína Wnt1/genética , Idoso , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinoma de Células Escamosas/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias dos Seios Paranasais/genética , Fatores de Transcrição/metabolismo , Proteína Wnt1/metabolismo , beta Catenina/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35086929

RESUMO

In Drosophila melanogaster, loss of regenerative capacity in wing imaginal discs coincides with an increase in systemic levels of the steroid hormone ecdysone, a key coordinator of their developmental progression. Regenerating discs release the relaxin hormone Dilp8 (Drosophila insulin-like peptide 8) to limit ecdysone synthesis and extend the regenerative period. Here, we describe how regenerating tissues produce a biphasic response to ecdysone levels: lower concentrations of ecdysone promote local and systemic regenerative signaling, whereas higher concentrations suppress regeneration through the expression of broad splice isoforms. Ecdysone also promotes the expression of wingless during both regeneration and normal development through a distinct regulatory pathway. This dual role for ecdysone explains how regeneration can still be completed successfully in dilp8- mutant larvae: higher ecdysone levels increase the regenerative activity of tissues, allowing regeneration to reach completion in a shorter time. From these observations, we propose that ecdysone hormone signaling functions to coordinate regeneration with developmental progression.


Assuntos
Ecdisona/metabolismo , Regeneração/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hormônios Esteroides Gonadais/metabolismo , Discos Imaginais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Larva/crescimento & desenvolvimento , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Asas de Animais/metabolismo , Proteína Wnt1/metabolismo
12.
Sci Adv ; 8(3): eabl5613, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35044823

RESUMO

De novo truncations in Interferon Regulatory Factor 2 Binding Protein Like (IRF2BPL) lead to severe childhood-onset neurodegenerative disorders. To determine how loss of IRF2BPL causes neural dysfunction, we examined its function in Drosophila and zebrafish. Overexpression of either IRF2BPL or Pits, the Drosophila ortholog, represses Wnt transcription in flies. In contrast, neuronal depletion of Pits leads to increased wingless (wg) levels in the brain and is associated with axonal loss, whereas inhibition of Wg signaling is neuroprotective. Moreover, increased neuronal expression of wg in flies is sufficient to cause age-dependent axonal loss, similar to reduction of Pits. Loss of irf2bpl in zebrafish also causes neurological defects with an associated increase in wnt1 transcription and downstream signaling. WNT1 is also increased in patient-derived astrocytes, and pharmacological inhibition of Wnt suppresses the neurological phenotypes. Last, IRF2BPL and the Wnt antagonist, CKIα, physically and genetically interact, showing that IRF2BPL and CkIα antagonize Wnt transcription and signaling.


Assuntos
Proteínas de Drosophila , Animais , Proteínas de Transporte/metabolismo , Criança , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Fator Regulador 2 de Interferon/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , Via de Sinalização Wnt , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
13.
Oncol Rep ; 47(2)2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34878149

RESUMO

C­terminal­binding protein 2 (CtBP2), a transcriptional co­repressor, plays a main role in tumorigenesis and in the development of multiple tumors. Transforming growth interacting factor (TGIF) is involved in a number of cellular signal transduction pathways and is related to tumor occurrence and development. In the present study, the proteins interacting with CtBP2 were identified and the mechanisms underlying the biological activity of CtBP2 in esophageal squamous cell carcinoma (ESCC) were investigated. The Search Tool for the Retrieval of Interacting Genes (STRING) database was used to search for known proteins interacting with CtBP2, and co­immunoprecipitation (Co­IP) assay was performed to validate the interactions. Reverse transcription­quantitative PCR (RT­qPCR), immunohistochemistry (IHC) and western blot analysis were performed to examine the expression levels of CtBP2 and TGIF in ESCC. The correlation between CtBP2 and TGIF was analyzed using Gene Expression Profiling Interactive Analysis (GEPIA) by Pearson's correlation analysis, and the co­localization of CtBP2 with TGIF in the ECA109 cells was identified using immunofluorescence staining. XAV939 treatment, CCK­8, 5­ethynyl­2'­deoxyuridine (EdU) staining, wound healing and Transwell assays were performed to investigate the signaling pathways involved in the biological activity of CtBP2 in ECA109 cells. According to the results obtained from STRING and Co­IP analysis, an interaction between CtBP2 and TGIF was indicated, and these proteins were co­localized in the nucleus. CtBP2 and TGIF mRNA and protein expression levels were robustly and simultaneously increased in both ESCC tissues and cell lines. There was a direct correlation between CtBP2 and TGIF expression levels in ESCC tissues, and both were significantly associated with metastasis and survival. The TGIF and CtBP2 expression levels were significantly increased or decreased simultaneously, in ECA109 cells transfected with LV­CtBP2 or sh­CtBP2, and vice versa. According to the results of CCK­8 assay, EdU staining and Transwell assay, CtBP2 promoted the proliferation, migration and invasion of ECA109 cells through the Wnt/ß­catenin pathway. On the whole, the present study demonstrates that CtBP2 interacts with TGIF and promotes the malignant progression of ESCC through the Wnt/ß­catenin pathway.


Assuntos
Oxirredutases do Álcool/genética , Proteínas Correpressoras/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Proteínas de Homeodomínio/genética , Proteínas Repressoras/genética , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Humanos
14.
Biochim Biophys Acta Mol Cell Res ; 1869(3): 119185, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34890713

RESUMO

Emerging evidences suggest abundant expression of Carboxy terminus of Hsc70 Interacting Protein or CHIP (alias STIP1 Homology and U-box Containing Protein 1 or STUB1) in colorectal carcinoma, but the mechanistic detail of this augmented expression pattern is unclear. The signature driver of canonical Wnt pathway, ß-catenin, and its co-activator RNA helicase p68, are also overexpressed in colorectal carcinoma. In this study, we describe a novel mechanism of Wnt/ß-catenin and p68 mediated transcriptional activation of CHIP gene leading to enhanced proliferation of colorectal carcinoma cells. Bioinformatic analyses reconfirmed an elevated CHIP expression level in colorectal carcinoma datasets. Wnt3A treatment and pharmacological activation of canonical Wnt signaling pathway resulted in increased nuclear translocation of ß-catenin, augmenting CHIP expression. Likewise, immunoblotting and Real time PCR following overexpression and knockdown of ß-catenin and p68 demonstrated upregulated and downregulated CHIP expression, respectively, at both mRNA and protein levels. p68 along with ß-catenin were found to occupy Transcription Factor 4 (TCF4) binding sites on endogenous CHIP promoter and regulate its transcription. After cloning CHIP promoter, the increased and decreased promoter activities of CHIP induced by overexpression and knockdown of either ß-catenin or p68 further confirmed transcriptional regulation of CHIP gene by Wnt/ß-catenin signaling cascade. Finally, enhanced cellular propagation and migration of colorectal carcinoma cells induced by 'Wnt/ß-catenin-p68-CHIP' axis established the significance of this pathway in oncogenesis. To the best of our knowledge, this is the first report elucidating the mechanistic details of transcriptional regulation of CHIP (STUB1) gene expression.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/patologia , RNA Helicases DEAD-box/metabolismo , Regulação Neoplásica da Expressão Gênica , Ubiquitina-Proteína Ligases/metabolismo , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Ciclo Celular , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , RNA Helicases DEAD-box/genética , Humanos , Regiões Promotoras Genéticas , Ativação Transcricional , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/genética , Proteína Wnt1/genética , beta Catenina/genética
15.
Cardiovasc Res ; 118(2): 573-584, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33576779

RESUMO

AIMS: Angiotensin (Ang) II signalling has been suggested to promote cardiac fibrosis in inflammatory heart diseases; however, the underlying mechanisms remain obscure. Using Agtr1a-/- mice with genetic deletion of angiotensin receptor type 1 (ATR1) and the experimental autoimmune myocarditis (EAM) model, we aimed to elucidate the role of Ang II-ATR1 pathway in development of heart-specific autoimmunity and post-inflammatory fibrosis. METHODS AND RESULTS: EAM was induced in wild-type (WT) and Agtr1a-/- mice by subcutaneous injections with alpha myosin heavy chain peptide emulsified in complete Freund's adjuvant. Agtr1a-/- mice developed myocarditis to a similar extent as WT controls at day 21 but showed reduced fibrosis and better systolic function at day 40. Crisscross bone marrow chimaera experiments proved that ATR1 signalling in the bone marrow compartment was critical for cardiac fibrosis. Heart infiltrating, bone-marrow-derived cells produced Ang II, but lack of ATR1 in these cells reduced transforming growth factor beta (TGF-ß)-mediated fibrotic responses. At the molecular level, Agtr1a-/- heart-inflammatory cells showed impaired TGF-ß-mediated phosphorylation of Smad2 and TAK1. In WT cells, TGF-ß induced formation of RhoA-GTP and RhoA-A-kinase anchoring protein-Lbc (AKAP-Lbc) complex. In Agtr1a-/- cells, stabilization of RhoA-GTP and interaction of RhoA with AKAP-Lbc were largely impaired. Furthermore, in contrast to WT cells, Agtr1a-/- cells stimulated with TGF-ß failed to activate canonical Wnt pathway indicated by suppressed activity of glycogen synthase kinase-3 (GSK-3)ß and nuclear ß-catenin translocation and showed reduced expression of Wnts. In line with these in vitro findings, ß-catenin was detected in inflammatory regions of hearts of WT, but not Agtr1a-/- mice and expression of canonical Wnt1 and Wnt10b were lower in Agtr1a-/- hearts. CONCLUSION: Ang II-ATR1 signalling is critical for development of post-inflammatory fibrotic remodelling and dilated cardiomyopathy. Our data underpin the importance of Ang II-ATR1 in effective TGF-ß downstream signalling response including activation of profibrotic Wnt/ß-catenin pathway.


Assuntos
Angiotensina II/metabolismo , Doenças Autoimunes/metabolismo , Autoimunidade , Linfócitos T CD4-Positivos/metabolismo , Miocardite/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Via de Sinalização Wnt , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/imunologia , Miocardite/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Receptor Tipo 1 de Angiotensina/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
16.
J Cancer Res Ther ; 17(6): 1404-1418, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34916371

RESUMO

BACKGROUND AND OBJECTIVES: Metabolic shifting from mitochondrial respiration to glycolysis characterizes malignant cells from its normal counterparts and is attributed to overactivation of oncogenic signaling pathways. Hence, this study intended to investigate the influence of canagliflozin (CAN) and/or γ-irradiation (γ-IR) on HepG2 cell proliferation, crosstalk between phosphatidylinositol 3-kinases (PI3K)/AKT/glycogen synthase kinase-3-ß (GSK3-ß)/mTOR and Wnt/ß-catenin signaling pathways, and their regulation of diverse processes, such as endoplasmic reticulum (ER) stress, autophagy, and apoptosis. MATERIALS AND METHODS: HepG2 cells were treated with different doses of CAN and then exposed to different doses of γ-IR to achieve optimization that was based on cytotoxicity and clonogenic assays, respectively. The effects of CAN and/or γ-IR on glycolytic metabolism, cellular bioenergetics, oxidative stress, ER stress and autophagy biomarkers, expression of PI3K/AKT/GSK3-ß/mTOR and Wnt/ß-Catenin signaling pathways, and apoptotic markers were monitored. RESULTS: CAN enhanced the antitumor potential of γ-IR as displayed by a significant inhibition of clonogenic survival in HepG2 cells via inhibition of glucose uptake, lactate release, and modulation of ER stress-mediated autophagy; switched it to apoptosis; as well as disabled signaling pathways which contribute to metabolic reprogramming and tumor progression induced by γ-IR that confer radioresistance and treatment failure. CONCLUSION: Our study sheds light on the effective combination of CAN and γ-IR in hepatocellular carcinoma treatment and necessitates CAN treatment prior to γ-IR to overcome metabolic reprogramming-associated radioresistance and improve curative outcomes.


Assuntos
Autofagia , Canagliflozina/farmacologia , Carcinoma Hepatocelular/patologia , Estresse do Retículo Endoplasmático , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Neoplasias Hepáticas/patologia , Apoptose , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Proliferação de Células , Quimiorradioterapia , Raios gama , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/terapia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Dis Markers ; 2021: 1405045, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790276

RESUMO

BACKGROUND: Growing studies have demonstrated that long noncoding RNAs (lncRNAs) play important roles in tumor progression. In this study, we aimed to explore the potential roles of lncRNA LINC00958 (LINC00958) and its biological functions in epithelial ovarian cancer (EOC). METHODS: The expression of LINC00958 in 11 cases of EOC and adjacent nontumor specimens and five cell lines was detected by qRT-PCR. CCK-8, colony formation, and flow cytometry assays were conducted to study the cell viabilities of EOC cells. Wound scratch and transwell analyses were carried out for the examination of cell invasion and migration of EOC cells. The targeting associations between LINC00958 and STAT1 were demonstrated by ChIP analyses combined with luciferase reporter assays. The related proteins of Wnt/ß-catenin signaling were determined using RT-PCR. RESULTS: Higher levels of LINC00958 were observed in EOC tissues and cell lines. Our data also revealed that high LINC00958 expression was partly induced by STAT1. Functionally, knockdown of LINC00958 suppressed the proliferation, migration, and invasion of EOC cells. Mechanistic investigation showed that the inhibitory effect of LINC00958 knockdown on EOC cells was mediated by the Wnt/ß-catenin signaling. CONCLUSION: Our findings suggested that STAT1-induced overexpression of LINC00958 promoted EOC progression by modulating Wnt/ß-catenin signaling.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Epitelial do Ovário/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/patologia , RNA Longo não Codificante/genética , Fator de Transcrição STAT1/metabolismo , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/metabolismo , Proliferação de Células , Feminino , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Prognóstico , Fator de Transcrição STAT1/genética , Células Tumorais Cultivadas , Proteína Wnt1/genética , beta Catenina/genética
18.
Biol Open ; 10(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34779478

RESUMO

Wnt signalling is a core pathway involved in a wide range of developmental processes throughout the metazoa. In vitro studies have suggested that the small GTP binding protein Arf6 regulates upstream steps of Wnt transduction, by promoting the phosphorylation of the Wnt co-receptor, LRP6, and the release of ß-catenin from the adherens junctions. To assess the relevance of these previous findings in vivo, we analysed the consequence of the absence of Arf6 activity on Drosophila wing patterning, a developmental model of Wnt/Wingless signalling. We observed a dominant loss of wing margin bristles and Senseless expression in Arf6 mutant flies, phenotypes characteristic of a defect in high level Wingless signalling. In contrast to previous findings, we show that Arf6 is required downstream of Armadillo/ß-catenin stabilisation in Wingless signal transduction. Our data suggest that Arf6 modulates the activity of a downstream nuclear regulator of Pangolin activity in order to control the induction of high level Wingless signalling. Our findings represent a novel regulatory role for Arf6 in Wingless signalling.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
19.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34681943

RESUMO

Glioblastoma (GBM) is an extremely aggressive brain tumor awaiting novel, efficient, and minimally toxic treatment. Curcuminoids (CCM), polyphenols from Curcuma longa, and sodium butyrate (NaBu), a histone deacetylase inhibitor naturally occurring in the human body, await elucidation as potential anti-GBM agents. Thus, the aim of this study was to analyze CCM and NaBu both separately and as a combination treatment using three GBM cell lines. MTT was used for cytotoxicity evaluation, and the combination index was calculated for synergism prediction. Cell cycle, apoptosis, and reactive oxygen species (ROS) generation were analyzed using flow cytometry. DNA methylation was verified by MS-HRM and mRNA expression by qPCR. The permeability through the blood-brain barrier (BBB) and through the nasal cavity was evaluated using PAMPA model. The results of this study indicate that CCM and NaBu synergistically reduce the viability of GBM cells inducing apoptosis and cell cycle arrest. These effects are mediated via ROS generation and changes in gene expression, including upregulation of Wnt/ß-catenin pathway antagonists, SFRP1, and RUNX3, and downregulation of UHRF1, the key epigenetic regulator. Moreover, NaBu ameliorated CCM permeability through the BBB and the nasal cavity. We conclude that CCM and NaBu are promising agents with anti-GBM properties.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Ácido Butírico/farmacologia , Diarileptanoides/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Apoptose , Ciclo Celular , Movimento Celular , Proliferação de Células , Quimioterapia Combinada , Glioblastoma/metabolismo , Glioblastoma/patologia , Antagonistas dos Receptores Histamínicos/farmacologia , Humanos , Células Tumorais Cultivadas , Proteína Wnt1/genética , beta Catenina/genética
20.
Oncogene ; 40(50): 6720-6735, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34657130

RESUMO

Epithelial-mesenchymal transition (EMT) refers to the acquisition of mesenchymal properties in cells participating in tumor progression. One hallmark of EMT is the increased level of active ß-catenin, which can trigger the transcription of Wnt-specific genes responsible for the control of cell fate. We investigated how Monocyte Chemotactic Protein-1-Induced Protein-1 (MCPIP1), a negative regulator of inflammatory processes, affects EMT in a clear cell renal cell carcinoma (ccRCC) cell line, patient tumor tissues and a xenotransplant model. We showed that MCPIP1 degrades miRNAs via its RNase activity and thus protects the mRNA transcripts of negative regulators of the Wnt/ß-catenin pathway from degradation, which in turn prevents EMT. Mechanistically, the loss of MCPIP1 RNase activity led to the upregulation of miRNA-519a-3p, miRNA-519b-3p, and miRNA-520c-3p, which inhibited the expression of Wnt pathway inhibitors (SFRP4, KREMEN1, CXXC4, CSNK1A1 and ZNFR3). Thus, the level of active nuclear ß-catenin was increased, leading to increased levels of EMT inducers (SNAI1, SNAI2, ZEB1 and TWIST) and, consequently, decreased expression of E-cadherin, increased expression of mesenchymal markers, and acquisition of the mesenchymal phenotype. This study revealed that MCPIP1 may act as a tumor suppressor that prevents EMT by stabilizing Wnt inhibitors and decreasing the levels of active ß-catenin and EMT inducers.


Assuntos
Carcinoma de Células Renais/patologia , Transição Epitelial-Mesenquimal , Fatores de Transcrição Forkhead/fisiologia , MicroRNAs/antagonistas & inibidores , Ribonucleases/metabolismo , Fatores de Transcrição/metabolismo , Proteína Wnt1/antagonistas & inibidores , beta Catenina/antagonistas & inibidores , Animais , Apoptose , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Movimento Celular , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Ribonucleases/genética , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...