Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Microvasc Res ; 143: 104400, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35724741

RESUMO

Accumulating evidences have pointed that foxc1a is essential for vascular development and integrity maintenance through regulating the expression of downstream genes and interacting with signaling pathways. However, the underling cellular and molecular mechanisms of foxc1a in regulating vascular development remain undetermined. Based on two different foxc1a mutant zebrafish lines (foxc1anju18 and foxc1anju19 which generated predicted truncated foxc1a proteins with 50aa and 315aa respectively), we found that around 30 % of foxc1anju18 zebrafish exhibited severe vascular developmental defects with obvious hemorrhage in hindbrain and trunk at embryonic stages. Confocal imaging analysis revealed that the formation of middle cerebral vein (MCeV), intra-cerebral central arteries (CtAs) and dorsal longitudinal vein (DLV) of brain vessels was significantly blocked in foxc1anju18enbryos. Injection of exogenous full length and foxc1anju19 truncated foxc1a mRNA both rescued the deficiency of foxc1anju18 embryos. Transcriptome analysis revealed 186 DEGs in foxc1anju18 zebrafish among which amotl2a and ctnnb1 expression were reduced and functionally associated with adherens junctions. Dual-Luciferase assays validated amotl2a and ctnnb1 were both directly transactivated by foxc1a. Rescue experiments demonstrated that amotl2a was mainly responsible for the vascular integrity caused by foxc1a mutation and also coordinated with ctnnb1 to regulate brain vascular development. Our data point to a novel clue that foxc1a regulates vascular integrity and brain vascular development through targeting amotl2a and ctnnb1.


Assuntos
Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Encéfalo/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
2.
Environ Toxicol ; 36(11): 2225-2235, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34323359

RESUMO

Angiomotin-like 2 (AMOTL2) is a key modulator of signaling transduction and participates in the regulation of various cellular progresses under diverse physiological and pathological conditions. However, whether AMOTL2 participates in asthma pathogenesis has not been fully studied. In the present work, we studied the possible role and mechanism of AMOTL2 in regulating transforming growth factor-ß1 (TGF-ß1)-induced proliferation and extracellular matrix (ECM) deposition of airway smooth muscle (ASM) cells. Our results showed marked reductions in the abundance of AMOTL2 in TGF-ß1-stimulated ASM cells. Cellular functional investigations confirmed that the up-regulation of AMOTL2 dramatically decreased the proliferation and ECM deposition induced by TGF-ß1 in ASM cells. In contrast, the depletion of AMOTL2 exacerbated TGF-ß1-induced ASM cell proliferation and ECM deposition. Further research revealed that the overexpression of AMOTL2 restrained the activation of Yes-associated protein 1 (YAP1) in TGF-ß1-stimulated ASM cells. Moreover, the reactivation of YAP1 markedly reversed AMOTL2-mediated suppression of TGF-ß1-induced ASM cell proliferation and ECM deposition. Together, these findings suggest that AMOTL2 restrains TGF-ß1-induced proliferation and ECM deposition of ASM cells by down-regulating YAP1 activation.


Assuntos
Proteínas de Transporte/genética , Matriz Extracelular , Miócitos de Músculo Liso , Fator de Crescimento Transformador beta1 , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proliferação de Células , Células Cultivadas , Regulação para Baixo , Camundongos , Miócitos de Músculo Liso/citologia , Fator de Crescimento Transformador beta1/farmacologia , Proteínas de Sinalização YAP
3.
Cancer Cell Int ; 20(1): 558, 2020 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33292274

RESUMO

BACKGROUND: Extracellular vesicles (EVs) secreted by tumours, including exosomes, are important factors that regulate cell-cell interactions in oncogenesis. Although EV studies are ongoing, the biological understanding of EV-miRNAs derived from brain tumour spheroid-forming cells (BTSCs) of medulloblastoma is poor. PURPOSES: We explored the specific cellular miRNAs and EV-miRNAs in medulloblastoma BTSCs to determine their potential biological function. METHODS: Bulk tumor cells (BTCs) and BTSCs were cultured under different conditions from medulloblastoma tissues (N = 10). RESULTS: Twenty-four miRNAs were simultaneously increased in both cells and EVs derived from BTSCs in comparison to BTCs. After inhibition of miR-135b or miR135a which were the most significantly increased in BTSCs, cell viability, self-renewal and stem cell marker expression decreased remarkably. Through integrated analysis of mRNAs and miRNAs data, we found that angiomotin-like 2 (AMOTL2), which was significantly decreased, was targeted by both miR-135b and miR-135a. STAT6 and GPX8 were targeted only by miR-135a. Importantly, low expression of AMOTL2 was significantly associated with overall poor survival in paediatric Group 3 and Group 4 medulloblastoma patients. CONCLUSION: Our results indicated that inhibition of miR-135b or miR-135a leads to suppress stemness of BTSC through modulation of AMOTL2.

4.
J Transl Med ; 16(1): 98, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29650031

RESUMO

Members in Motin family, or Angiomotins (AMOTs), are adaptor proteins that localize in the membranous, cytoplasmic or nuclear fraction in a cell context-dependent manner. They control the bioprocesses such as migration, tight junction formation, cell polarity, and angiogenesis. Emerging evidences have demonstrated that AMOTs participate in cancer initiation and progression. Many of the previous studies have focused on the involvement of AMOTs in Hippo-YAP1 pathway. However, it has been controversial for years that AMOTs serve as either positive or negative growth regulators in different cancer types because of the various cellular origins. The molecular mechanisms of these opposite roles of AMOTs remain elusive. This review comprehensively summarized how AMOTs function physiologically and how their dysregulation promotes or inhibits tumorigenesis. Better understanding the functional roles of AMOTs in cancers may lead to an improvement of clinical interventions as well as development of novel therapeutic strategies for cancer patients.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Proteínas de Membrana/metabolismo , Animais , Desenvolvimento Embrionário , Humanos , Proteínas de Membrana/química , Modelos Biológicos , Neovascularização Fisiológica , Transdução de Sinais
5.
Adv Anat Embryol Cell Biol ; 229: 47-68, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29177764

RESUMO

In placental mammalian development, the first cell differentiation produces two distinct lineages that emerge according to their position within the embryo: the trophectoderm (TE, placenta precursor) differentiates in the surface, while the inner cell mass (ICM, fetal body precursor) forms inside. Here, we discuss how such position-dependent lineage specifications are regulated by the RHOA subfamily of small GTPases and RHO-associated coiled-coil kinases (ROCK). Recent studies in mouse show that activities of RHO/ROCK are required to promote TE differentiation and to concomitantly suppress ICM formation. RHO/ROCK operate through the HIPPO signaling pathway, whose cell position-specific modulation is central to establishing unique gene expression profiles that confer cell fate. In particular, activities of RHO/ROCK are essential in outside cells to promote nuclear localization of transcriptional co-activators YAP/TAZ, the downstream effectors of HIPPO signaling. Nuclear localization of YAP/TAZ depends on the formation of apicobasal polarity in outside cells, which requires activities of RHO/ROCK. We propose models of how RHO/ROCK regulate lineage specification and lay out challenges for future investigations to deepen our understanding of the roles of RHO/ROCK in preimplantation development. Finally, as RHO/ROCK may be inhibited by certain pharmacological agents, we discuss their potential impact on human preimplantation development in relation to fertility preservation in women.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Quinases Associadas a rho , Animais , Blastocisto , Diferenciação Celular , Embrião de Mamíferos , Feminino , Camundongos , Gravidez , Transdução de Sinais , Quinases Associadas a rho/metabolismo
6.
EMBO Rep ; 17(1): 64-78, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26598551

RESUMO

LATS1/2 (large tumor suppressor) kinases and the Angiomotin family proteins are potent inhibitors of the YAP (yes-associated protein) oncoprotein, but the underlying molecular mechanism is not fully understood. Here, we report for the first time that USP9X is a deubiquitinase of Angiomotin-like 2 (AMOTL2) and that AMOTL2 mono-ubiquitination is required for YAP inhibition. USP9X knockdown increased the LATS-mediated phosphorylation of YAP and decreased the transcriptional output of YAP. Conversely, over-expression of USP9X reactivated YAP in densely cultured cells. Both genetic and biochemical approaches identified AMOTL2 as a target of USP9X. AMOTL2 was found to be ubiquitinated at K347 and K408, which both reside in the protein's coiled-coil domain. The AMOTL2 K347/408R mutant, which cannot be ubiquitinated, was impaired in its ability to inhibit YAP. Furthermore, ubiquitinated AMOTL2 can bind to the UBA domain of LATS kinase, and this domain is required for the function of LATS. Our results provide novel insights into the activation mechanisms of core Hippo pathway components.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Angiomotinas , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Proteínas dos Microfilamentos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitinação , Proteínas de Sinalização YAP
7.
Int J Mol Sci ; 18(4)2017 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-28441737

RESUMO

Scaffold proteins play pivotal roles in the regulation of signaling pathways, integrating external and internal stimuli to various cellular outputs. We report the pattern of cellular and subcellular expression of scaffoldins angiomotin-like 2 (AmotL2), FK506 binding protein 5 (FKBP51) and IQ motif containing GTPase-activating protein 1 (IQGAP1) in colorectal cancer (CRC) and metastases in liver resected after oxaliplatin-based chemotherapy (CT). Positive immunostaining for the three scaffoldins was found in most cells in healthy colon, tumor, healthy liver and metastasized liver. The patterns of expression of AmotL2, FKBP51 and IQGAP1 show the greatest variability in immune system cells and neurons and glia cells and the least in blood vessel cells. The simultaneous subcellular localization in tumor cells and other cell types within the tumor suggest an involvement of these three scaffoldins in cancer biology, including a role in Epithelial Mesenchymal Transition. The display in differential localization and quantitative expression of AmotL2, FKBP51, and IQGAP1 could be used as biomarkers for more accurate tumor staging and as potential targets for anti-cancer therapeutics by blocking or slowing down their interconnecting functions. Tough further research needs to be done in order to improve these assessments.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Compostos Organoplatínicos/uso terapêutico , Angiomotinas , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Proteínas de Transporte/metabolismo , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Humanos , Imuno-Histoquímica , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Microscopia de Fluorescência , Oxaliplatina , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
8.
J Biol Chem ; 290(32): 19387-401, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25998128

RESUMO

The mechanistic target of rapamycin (mTOR) and Hippo signaling pathways are two major signaling cascades that coordinately regulate cell growth and proliferation. Dysregulation of these pathways plays a critical role in gliomagenesis. Recent reports have provided evidence of cross-talk between the mTOR and Hippo pathways; however, a complete description of the signaling relationships between these pathways remains to be elucidated. Utilizing a gene-trapping strategy in a mouse glioma model, we report the identification of AMOTL2 as a candidate substrate for mTORC2. AMOTL2 is phosphorylated at serine 760 by mTORC2. Mutation of AMOTL2 mimicking constitutive Ser(760) phosphorylation blocks its ability to bind and repress YAP leading to increased relative expression of known YAP gene targets. Moreover, overexpression of AMOTL2 or a nonphosphorylatable AMOTL2-S760A mutant inhibited YAP-induced transcription, foci formation, growth, and metastatic properties, whereas overexpression of a phosphomimetic AMOTL2-S760E mutant negated these repressive effects of AMOTL2 in glioblastoma (GBM) cells in vitro. Similar effects on xenograft growth were observed in GBM cells expressing these AMOTL2 Ser(760) mutants. YAP was also shown to be required for Rictor-mediated GBM growth and survival. Finally, an analysis of mTORC2/AMOTL2/YAP activities in primary GBM samples supported the clinical relevance of this signaling cascade, and we propose that pharmacological agents cotargeting these regulatory circuits may hold therapeutic potential.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias Encefálicas/metabolismo , Proteínas de Transporte/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Complexos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Angiomotinas , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/genética , Mutação , Invasividade Neoplásica , Transplante de Neoplasias , Fosfoproteínas/genética , Fosforilação , Proteína Companheira de mTOR Insensível à Rapamicina , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Fatores de Transcrição , Transplante Heterólogo , Proteínas de Sinalização YAP
9.
J Exp Clin Cancer Res ; 43(1): 107, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594748

RESUMO

BACKGROUND: Tumor cells have the ability to invade and form small clusters that protrude into adjacent tissues, a phenomenon that is frequently observed at the periphery of a tumor as it expands into healthy tissues. The presence of these clusters is linked to poor prognosis and has proven challenging to treat using conventional therapies. We previously reported that p60AmotL2 expression is localized to invasive colon and breast cancer cells. In vitro, p60AmotL2 promotes epithelial cell invasion by negatively impacting E-cadherin/AmotL2-related mechanotransduction. METHODS: Using epithelial cells transfected with inducible p60AmotL2, we employed a phenotypic drug screening approach to find compounds that specifically target invasive cells. The phenotypic screen was performed by treating cells for 72 h with a library of compounds with known antitumor activities in a dose-dependent manner. After assessing cell viability using CellTiter-Glo, drug sensitivity scores for each compound were calculated. Candidate hit compounds with a higher drug sensitivity score for p60AmotL2-expressing cells were then validated on lung and colon cell models, both in 2D and in 3D, and on colon cancer patient-derived organoids. Nascent RNA sequencing was performed after BET inhibition to analyse BET-dependent pathways in p60AmotL2-expressing cells. RESULTS: We identified 60 compounds that selectively targeted p60AmotL2-expressing cells. Intriguingly, these compounds were classified into two major categories: Epidermal Growth Factor Receptor (EGFR) inhibitors and Bromodomain and Extra-Terminal motif (BET) inhibitors. The latter consistently demonstrated antitumor activity in human cancer cell models, as well as in organoids derived from colon cancer patients. BET inhibition led to a shift towards the upregulation of pro-apoptotic pathways specifically in p60AmotL2-expressing cells. CONCLUSIONS: BET inhibitors specifically target p60AmotL2-expressing invasive cancer cells, likely by exploiting differences in chromatin accessibility, leading to cell death. Additionally, our findings support the use of this phenotypic strategy to discover novel compounds that can exploit vulnerabilities and specifically target invasive cancer cells.


Assuntos
Neoplasias do Colo , Mecanotransdução Celular , Humanos , Linhagem Celular Tumoral , Detecção Precoce de Câncer , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética
10.
Cells ; 12(13)2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37443716

RESUMO

The spread of tumor cells and the formation of distant metastasis remain the main causes of mortality in cancer patients. However, the mechanisms governing the release of cells from micro-environmental constraints remain unclear. E-cadherin negatively controls the invasion of epithelial cells by maintaining cell-cell contacts. Furthermore, the inactivation of E-cadherin triggers invasion in vitro. However, the role of E-cadherin is complex, as metastasizing cells maintain E-cadherin expression, which appears to have a positive role in the survival of tumor cells. In this report, we present a novel mechanism delineating how E-cadherin function is modulated to promote invasion. We have previously shown that E-cadherin is associated with p100AmotL2, which is required for radial actin formation and the transmission of mechanical force. Here, we present evidence that p60AmotL2, which is expressed in invading tumor cells, binds to the p100AmotL2 isoform and uncouples the mechanical constraint of radial actin filaments. We show for the first time that the coupling of E-cadherin to the actin cytoskeleton via p100AmotL2 is directly connected to the nuclear membrane. The expression of p60AmotL2 inactivates this connection and alters the properties of the nuclear lamina, potentiating the invasion of cells into micropores of the extracellular matrix. In summary, we propose that the balance of the two AmotL2 isoforms is important in the modulation of E-cadherin function and that an imbalance of this axis promotes ameboid cell invasion.


Assuntos
Amoeba , Humanos , Amoeba/metabolismo , Caderinas/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Células Epiteliais/metabolismo
11.
Cells ; 11(9)2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35563891

RESUMO

The immunophilin FKBP51, the angiomotin AmotL2, and the scaffoldin IQGAP1 are overexpressed in many types of cancer, with the highest increase in leucocytes from patients undergoing oxaliplatin chemotherapy. Inflammation is involved in the pathogenesis of nephrotoxicity induced by platinum analogs. Cilastatin prevents renal damage caused by cisplatin. This functional and confocal microscopy study shows the renal focal-segmental expression of TNFα after cisplatin administration in rats, predominantly of tubular localization and mostly prevented by co-administration of cilastatin. FKBP51, AmotL2 and IQGAP1 protein expression increases slightly with cilastatin administration and to a much higher extent with cisplatin, in a cellular- and subcellular-specific manner. Kidney tubule cells expressing FKBP51 show either very low or no expression of TNFα, while cells expressing TNFα have low levels of FKBP51. AmotL2 and TNFα seem to colocalize and their expression is increased in tubular cells. IQGAP1 fluorescence increases with cilastatin, cisplatin and joint cilastatin-cisplatin treatment, and does not correlate with TNFα expression or localization. These data suggest a role for FKBP51, AmotL2 and IQGAP1 in cisplatin toxicity in kidney tubules and in the protective effect of cilastatin through inhibition of dehydropeptidase-I.


Assuntos
Cilastatina , Cisplatino , Angiomotinas , Animais , Proteínas de Transporte/metabolismo , Cilastatina/metabolismo , Cilastatina/farmacologia , Cilastatina/uso terapêutico , Cisplatino/metabolismo , Cisplatino/toxicidade , Humanos , Ratos , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
12.
Int J Clin Exp Pathol ; 15(5): 206-214, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35698637

RESUMO

Hepatocellular carcinoma (HCC) is one of the deadliest malignancies in the world. There is a lack of effective treatment. Previous studies have shown that myocyte enhancer factor 2D (MEF2D) promotes the progression of HCC. Underlying mechanisms have not been fully elucidated. In this study, we reported experimental results obtained using double luciferase. Our results showed that AMOTL2, a negative regulator of Hippo/YAP signaling, and the MEF2 cis-acting element in the upstream region of its promoter bind to MEF2D, inhibiting its transcriptional expression. Studies confirmed that MEF2D affected the protein expression level of AMOTL2 and the YAP signaling activation. It promoted the migration and proliferation of hepatoma cells. We found that luteolin, a natural flavonoid, has anti-tumor activity in HCC cells by affecting YAP signaling transduction. In conclusion, we demonstrated that AMOTL2/YAP signaling is associated with MEF2D-related HCC progression. Luteolin is a promising anti-HCC compound for regulating this signaling.

13.
J Histochem Cytochem ; 70(1): 9-16, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34165350

RESUMO

Glioma stem cells (GSCs) live in a continuous process of stemness reprogramming to achieve specific cell commitment within the so-called GSC niches, specifically located in periarteriolar regions. In this review, we analyze the expression levels, cellular and subcellular location, and role of three scaffold proteins (IQGAP1, FKBP51, and AmotL2) in GSC niches. Scaffold proteins contribute to cell differentiation, migration, and angiogenesis in glioblastoma. It could be of diagnostic interest for establishing stages, for therapeutic targets, and for improving glioblastoma prognosis, which is still at the experimental level.


Assuntos
Angiomotinas/genética , Glioblastoma/genética , Proteínas de Ligação a Tacrolimo/genética , Proteínas Ativadoras de ras GTPase/genética , Diferenciação Celular , Glioblastoma/diagnóstico , Glioblastoma/tratamento farmacológico , Humanos , Células-Tronco Neoplásicas
14.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118858, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32950569

RESUMO

Protein phosphatase 2A (PP2A) complex comprises an extended family of intracellular protein serine/threonine phosphatases, that participate in different signaling transduction pathways. Different functions of PP2As are determined by the variety of regulatory subunits. In this study, CRISPR/Cas9-mediated loss-of-function screen revealed that PPP2R2A downregulation suppressed cell growth in NSCLC cells. AMOTL2 was identified and confirmed as a novel binding partner of PPP2R2A in NSCLC cells by mass spectrometry, CO-IP, GST pull-down and immunofluorescence. Upregulation of AMOTL2 also led to cell proliferation delay in human and mouse lung tumor cells. The proto-oncogene JUN is a key subunit of activator protein-1 (AP-1) transcription factor which plays crucial role in regulating tumorigenesis and its activity is negatively regulated by the phosphorylation at T239. Our results showed that either AMOTL2 upregulation or PPP2R2A downregulation led to great increase in JUN T239 phosphorylation. AMOTL2 bound PPP2R2A in cytoplasm, which reduced nuclear localization of PPP2R2A. In conclusion, AMOTL2 and PPP2R2A act respectively as negative and positive regulator of cell growth in NSCLC cells and function in the AMOTL2-PPP2R2A-JUN axis, in which AMOTL2 inhibits the entry of PPP2R2A into the nucleus to dephosphorylate JUN at T239.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas de Transporte/genética , Proteína Fosfatase 2/genética , Proteínas Proto-Oncogênicas c-jun/genética , Angiomotinas , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Fosforilação/genética , Proto-Oncogene Mas , Fator de Transcrição AP-1/genética , Regulação para Cima
15.
Aging (Albany NY) ; 13(5): 7382-7396, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33658392

RESUMO

Exosomes play important roles in the regulation of various processes in the tumor microenvironment. In this study, we explored the mechanisms of exosomal miR-149-5p in the pathogenesis of lung adenocarcinoma. Raw data were downloaded and normalized using the R package. Significantly expressed exosomal miRNAs were subjected to co-expression network analysis. The proliferation and apoptotic abilities of tumor cells were assessed by the proliferation and apoptosis assays. Univariate and multivariate analyses were performed to identify the independent risk factors of exosomal miR-149-5p and AMOTL2. Results showed that exosomal miR-149-5p was enriched in peripheral serum and tumor cells. The upregulation of exosomal miR-149-5p promoted the growth of tumor cells and inhibited apoptosis of tumor cells. Notably, AMOTL2, the target gene of exosomal miR-149-5p, was significantly downregulated in lung adenocarcinoma and may be considered as an independent risk factor of poor survival. In lung adenocarcinoma cells, AMOTL2 downregulation reversed the promoting effect of miR-149-5p on A549 cells growth and the inhibition effect of miR-149-5p on A549 cells apoptosis. Collectively, these results provide specific insights for further mechanistic studies on lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/genética , Exossomos/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Angiomotinas , Proteínas de Transporte/metabolismo , Proliferação de Células/genética , Predisposição Genética para Doença/genética , Humanos , Regulação para Cima
16.
J Histochem Cytochem ; 67(7): 481-494, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30794467

RESUMO

Glioblastoma (GB) is the most frequently occurring and aggressive primary brain tumor. Glioma stem cells (GSCs) and astrocytoma cells are the predominant malignant cells occurring in GB besides a highly heterogeneous population of migrating, neovascularizing and infiltrating myeloid cells that forms a complex tumor microenvironment (TME). Cross talk between the TME cells is pivotal in the biology of this tumor and, consequently, adaptor proteins at critical junctions of signaling pathways may be crucial. Scaffold proteins (scaffolins or scaffoldins) integrate external and internal stimuli to regulate various signaling pathways, interacting simultaneously with multiple proteins involved. We investigated by double and triple immunofluorescence the localization of IQGAP1, AmotL2, and FKBP51, three closely related scaffoldins, in malignant cells and TME of human GB tumors. We found that IQGAP1 is preferentially expressed in astrocytoma cells, AmotL2 in GSCs, and FKBP51 in white blood cells in human GB tumors. As GSCs are specially the target for novel therapies, we will investigate in further studies whether AmotL2 inhibition is effective in the treatment of GB.


Assuntos
Proteínas de Transporte/metabolismo , Glioblastoma/patologia , Proteínas de Ligação a Tacrolimo/metabolismo , Microambiente Tumoral , Proteínas Ativadoras de ras GTPase/metabolismo , Angiomotinas , Linhagem Celular Tumoral , Humanos , Espaço Intracelular/metabolismo , Transdução de Sinais
17.
Elife ; 4: e11375, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26473744

RESUMO

Several signaling pathways work together, via a protein called Amotl2a, to establish the size and shape of a zebrafish sense organ primordium.


Assuntos
Proteínas de Membrana/metabolismo , Órgãos dos Sentidos/embriologia , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Angiomotinas , Animais , Modelos Biológicos
18.
Elife ; 4: e08201, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26335201

RESUMO

During development, proliferation must be tightly controlled for organs to reach their appropriate size. While the Hippo signaling pathway plays a major role in organ growth control, how it senses and responds to increased cell density is still unclear. In this study, we use the zebrafish lateral line primordium (LLP), a group of migrating epithelial cells that form sensory organs, to understand how tissue growth is controlled during organ formation. Loss of the cell junction-associated Motin protein Amotl2a leads to overproliferation and bigger LLP, affecting the final pattern of sensory organs. Amotl2a function in the LLP is mediated together by the Hippo pathway effector Yap1 and the Wnt/ß-catenin effector Lef1. Our results implicate for the first time the Hippo pathway in size regulation in the LL system. We further provide evidence that the Hippo/Motin interaction is essential to limit tissue size during development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Angiomotinas , Animais , Proliferação de Células , Células Epiteliais/fisiologia , Proteínas de Sinalização YAP , Peixe-Zebra/genética
19.
Tissue Barriers ; 2(1): e28127, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24843842

RESUMO

The Hippo signaling pathway regulates a number of cellular events, including the control of cell fates in preimplantation mouse embryos. The inner and outer cells of the embryo show high and low levels of Hippo signaling, respectively. This position-dependent Hippo signaling promotes the specification of distinct cell fates. In a recent paper, we identified the molecular mechanism that controls Hippo signaling in preimplantation embryos. The junction-associated scaffold protein Angiomotin (Amot) plays a key role in this mechanism. At the adherens junctions of the inner cells, Amot activates the Hippo pathway by recruiting and activating the protein kinase large tumor suppressor (Lats). In contrast, Amot at the apical membrane of the outer cells suppresses Hippo signaling by interacting with F-actin. The phosphorylation of Amot inhibits its interaction with F-actin and activates Hippo signaling. We propose that Amot acts as a molecular switch for the Hippo pathway and links F-actin with Lats activity.

20.
Eur J Cell Biol ; 93(10-12): 478-85, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25012928

RESUMO

Podosomes are adhesive, matrix remodeling organelles that have been described in numerous cell types, including all three vertebrate muscle cell lineages. Podosomes have been intensively studied in smooth muscle cells, but they have also been described in cardiac myocytes and skeletal muscle cells where they are proposed to play a role in developmental remodeling of neuromuscular junction postsynaptic machinery. In this review, we summarize the current state of knowledge of podosomes in muscle cells, with a focus on their potential function at the maturing synapse.


Assuntos
Extensões da Superfície Celular/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miócitos de Músculo Liso/metabolismo , Junção Neuromuscular/metabolismo , Animais , Fusão Celular , Extensões da Superfície Celular/ultraestrutura , Humanos , Fibras Musculares Esqueléticas/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Miócitos de Músculo Liso/ultraestrutura , Junção Neuromuscular/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA